WO1998048837A9 - Polyalkylene oxide-modified single chain polypeptides - Google Patents

Polyalkylene oxide-modified single chain polypeptides

Info

Publication number
WO1998048837A9
WO1998048837A9 PCT/US1998/008654 US9808654W WO9848837A9 WO 1998048837 A9 WO1998048837 A9 WO 1998048837A9 US 9808654 W US9808654 W US 9808654W WO 9848837 A9 WO9848837 A9 WO 9848837A9
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
antigen
binding
chain
polyalkylene oxide
Prior art date
Application number
PCT/US1998/008654
Other languages
French (fr)
Other versions
WO1998048837A1 (en
Inventor
Marc Whitlow
Robert G L Shorr
David R Filpula
Lihsyng S Lee
Original Assignee
Enzon Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Enzon Inc filed Critical Enzon Inc
Priority to CA2288994A priority Critical patent/CA2288994C/en
Priority to AU72666/98A priority patent/AU7266698A/en
Priority to JP54734798A priority patent/JP2002505574A/en
Priority to EP98920001A priority patent/EP0979102A4/en
Publication of WO1998048837A1 publication Critical patent/WO1998048837A1/en
Publication of WO1998048837A9 publication Critical patent/WO1998048837A9/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • G01N33/6857Antibody fragments
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to the chemical modification of single chain polypeptides by means of covalent attachment of strands of poly(ethylene glycol) PEG and similar poly(alkylene oxides) to single chain polypeptide binding molecules that have the three dimensional folding and, thus, the binding ability and specificity, of the variable region of an antibody.
  • Such preparations of modified single chain polypeptide binding molecules have reduced immugenicity and antigenicity as well as having a longer halflife in the bloodstream as compared to the parent polypeptide. These beneficial properties of the modified single chain polypeptide binding molecules make them very useful in a variety of therapeutic applications.
  • the invention also relates to multivalent antigen-binding molecules capable of PEGylation. Compositions of, genetic constructions for, methods of use, and methods for producing PEGylated antigen-binding proteins are disclosed.
  • Antibodies are proteins generated by the immune system to provide a specific molecule capable of complexing with an invading molecule, termed an antigen. Natural antibodies have two identical antigen-binding sites, both of which are specific to a particular antigen. The antibody molecule "recognizes" the antigen by complexing its antigen-binding sites with areas of the antigen termed epitopes. The epitopes fit into the conformational architecture of the antigen- binding sites of the antibody, enabling the antibody to bind to the antigen.
  • the antibody molecule is composed of two identical heavy and two identical light polypeptide chains, held together by interchain disulfide bonds.
  • the remainder of this discussion on antibodies will refer only to one pair of light/heavy chains, as each light/heavy pair is identical.
  • Each individual light and heavy chain folds into regions of approximately 110 amino acids, assuming a conserved three- dimensional conformation.
  • the light chain comprises one variable region (V L ) and one constant region (C L ), while the heavy chain comprises one variable region (N H ) and three constant regions (C H 1, C H 2 and C H 3). Pairs of regions associate to form discrete structures.
  • the light and heavy chain variable regions associate to form an "Fv" area which contains the antigen-binding site.
  • the constant regions are not necessary for antigen binding and in some cases can be separated from the antibody molecule by proteolysis, yielding biologically active (i.e., binding) variable regions composed of half of a light chain and one quarter of a heavy chain.
  • a multivalent antigen-binding protein has more than one antigen-binding site.
  • a multivalent antigen-binding protein comprises two or more single-chain protein molecules.
  • Ladner et al. also discloses the use of the single chain antigen binding molecules in diagnostics, therapeutics, in vivo and in vitro imaging, purifications, and biosensors.
  • the use of the single chain antigen binding molecules in immobilized form, or in detectably labeled forms is also disclosed, as well as conjugates of the single chain antigen binding molecules with therapeutic agents, such as drugs or specific toxins, for delivery to a specific site in an animal, such as a human patient.
  • Huston et al. discloses a family of synthetic proteins having affinity for preselected antigens.
  • the contents of U.S.Patent 5,091,513 are incorporated by reference herein.
  • the proteins are characterized by one or more sequences of amino acids constituting a region that behaves as a biosynthetic antibody binding site (BABS).
  • the sites comprise (1 ) noncovalently associated or disulfide bonded synthetic V H and V L regions, (2) N H -V L or V L -N H single chains wherein the V H and V L are attached to a polypeptide linker, or (3) individual V H or V L domains.
  • the binding domains comprises complementarity determining regions (CDRs) linked to framework regions (FRs), which may be derived from separate immunoglobulins.
  • U.S. Patent 5,091,513 also discloses that three subregions (the CDRs) of the variable domain of each of the heavy and light chains of native immunoglobulin molecules collectively are responsible for antigen recognition and binding. These CDRs consist of one of the hypervariable regions or loops and of selected amino acids or amino acid sequences disposed in the framework regions that flank that particular hypervariable region. It is said that framework regions from diverse species are effective in maintaining CDRs from diverse other species in proper conformation so as to achieve true immunochemical binding properties in a biosynthetic protein.
  • U.S. Patent 5 ,091 ,513 includes a description of a chimeric polypeptide that is a single chain composite polypeptide comprising a complete antibody binding site.
  • This single chain composite polypeptide is described as having a structure patterned after tandem V H and V L domains, with a carboxyl terminal of one attached through an amino acid sequence to the amino terminal of the other. It thus comprises an amino acid sequence that is homologous to a portion of the variable region of an immunoglobulin heavy chain (N H ) peptide bonded to a second amino acid sequence that was homologous to a portion of the variable region of an immunoglobulin light chain (V L ).
  • amino acid sequences such as the single chain polypeptides described above, and fusion proteins thereof, have not been associated with significant antigenicity in mammals, it has been desirable to prolong the circulating life and even further reduce the possibility of an antigenic response.
  • the relatively small size of the polypeptides and their delicate structure/activity relationship have made polyethylene glycol modification difficult and unpredictable. Most importantly, it was unknown how to modulate retained activity of the polypeptides after conjugation with polymers, such as PEG.
  • PEG polyethylene glycol
  • PEG polyalkalene oxide
  • the hydroxyl end groups of the polymer must first be converted into reactive functional groups. This process is frequently referred to as “activation” and the product is called “activated PEG” or activated polyalkylene oxide.
  • activated PEG Methoxy poly(ethylene glycol) (mPEG), capped on one end with a functional group, reactive towards amines on a protein molecule, is used in most cases.
  • the activated polymers are reacted with a therapeutic agent having nucleophilic functional groups that serve as attachment sites.
  • nucleophilic functional group commonly used as an attachment site is the e-amino groups of lysines.
  • Free carboxylic acid groups suitably activated carbonyl groups, oxidized carbohydrate moieties and mercapto groups have also been used as attachment sites.
  • PEGs such as succinimidyl succinate derivatives of PEG (“SS-PEG”)
  • SS-PEG succinimidyl succinate derivatives of PEG
  • Zalipsky in U.S. Patent No.5, 122,614, discloses poly(ethylene glycol)-N- succinimide carbonate and its preparation. This form of the polymer is said to react readily with the amino groups of proteins, as well as low molecular weight peptides and other materials that contain free amino groups.
  • linkages between the amino groups of the protein, and the PEG are also known in the art, such as urethane linkages (Veronese et al, Appl. Biochem. Biotechnol 77:141-152 (1985)), carbamate linkages (Beauchamp et al, Analyt. Biochem. 131:25-33 (1983)), and others.
  • IgG immunoglobulin G
  • poly(ethylene glycol) that has previously been activated by cyanuric chloride.
  • the coupled IgG was studied for physicochemical and biological properties such as molecular structure, size- exclusion chromatographic behavior, surface activity, interfacial aggregability, heat aggregability inducing nonspecific complement activation, and antigen- binding activity.
  • the poly(ethylene glycol) coupling to IgG increased the apparent Stokes' radius and the surface activity of IgG and stabilized IgG on heating and/or on exposure to interfaces, while no structural denaturation of IgG was observed.
  • the suppressed nonspecific aggregability was interpreted mainly by difficulty in association between the modified IgG molecules.
  • Tullis in U.S. Patent No.4,904,582, describes oligonucleotide conjugates wherein the oligonucleotides are joined through a linking arm to a hydrophobic moiety, which could be a polyalkyleneoxy group.
  • the resulting conjugates are said to be more efficient in membrane transport, so as to be capable of crossing the membrane and effectively modulating a transcriptional system.
  • the compositions can be used in vitro and in vivo, for studying cellular processes, protecting mammalian hosts from pathogens, and the like.
  • Benhar et al. replaced these two lysine residues with arginine residues and were able to obtain an active immunotoxin that was 3-fold more resistant to inactivation by derivatization.
  • aAnother suggestion for overcoming these problems discussed above is to use longer, higher molecular weight polymers. These materials, however, are difficult to prepare and expensive to use. Further, they provide little improvement over more readily available polymers.
  • the present invention relates to polyalkylene oxide/amino acid sequence conjugates and processes for preparing them.
  • Suitable amino acid sequences are peptides, such as, single chain polypeptides having binding affinity for an antigen, for example, those described by Ladner et al. in U.S. Patent No. 4,946,778 and Huston et al. in U.S. Patent No. 5,091,513.
  • the present invention relates to a physiologically active, substantially non-immunogenic polypeptide conjugate containing at least one polyalkylene oxide strand coupled to a single chain polypeptide having binding affinity for an antigen.
  • the single chain polypeptide includes:
  • the present invention relates to a process for preparing physiologically active, substantially non-immunogenic polypeptide compositions.
  • the process includes coupling a polyalkylene oxide to a single chain polypeptide having the attributes described above.
  • the poly(alkylene oxides) used herein are poly(ethylene glycols) that have been activated for coupling to the target polypeptide.
  • the invention is also directed to a single-chain antigen-binding polypeptide
  • a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within a range of about one-fold to about ten-fold of the antigen binding affinity of the native, unconj ugated foim of the single-chain antigen-binding polypeptide.
  • the invention is also directed to a single-chain antigen-binding polypeptide -polyalkylene oxide conjugate, comprising:
  • a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about ten-fold of the antigen binding affinity of the native, unconjugated form of the single-chain antigen-binding polypeptide.
  • the invention is also directed to a single-chain antigen-binding polypeptide -polyalkylene oxide conjugate, comprising:
  • the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about five-fold of the antigen binding affinity of the native, unconjugated form of the single-chain antigen-binding polypeptide.
  • the invention is also directed to a single-chain antigen-binding polypeptide
  • a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about two-fold of the antigen binding affinity of the native, unconjugated form of the single-chain antigen-binding polypeptide.
  • the invention is also directed to a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
  • a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue wherein the Cys residue is capable of polyalkylene oxide conjugation and the Cys residue is located at a position selected from the group consisting of (i) the amino acid position 11 , 12, 13 , 14 or 15 of the light chain variable region; (ii) the amino acid position 77, 78 or 79 of the light chain variable region; (iii) the amino acid position 11, 12, 13, 14 or 15 of the heavy chain variable region; (iv) the amino acid position 82B, 82C or 83 of the heavy chain variable region; (v) any amino acid position of the peptide linker; (vi) adjacent to the C-terminus of polypeptide
  • the invention is also directed to a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
  • the single-chain antigen-binding polypeptide has at least three consecutive Lys residues wherein the consecutive Lys residues are capable of polyalkylene oxide conjugation and any one of the consecutive Lys residues is located at a position selected from the group consisting of (i) any amino acid position of the peptide linker; (ii) adjacent to the C-terminus of polypeptide (a) or (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
  • SCA sFv
  • oligo-lysine sFv generate a "hot spot" for polyalkylene oxide conjugation.
  • the invention is also directed to a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising: (a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain;
  • a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least two consecutive Cys residue wherein the consecutive Cys residues are capable of polyalkylene oxide conjugation and any one of the consecutive Cys residues is located at a position selected from the group consisting of (i) any amino acid position of the peptide linker; (ii) adjacent to the C-terminus of polypeptide (a) or (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
  • the invention is further directed to a genetic sequence encoding a single- chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
  • a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue wherein the Cys residue is capable of polyalkylene oxide conjugation and the Cys residue is located at a position selected from the group consisting of (i) the amino acid position 11 , 12, 13, 14 or 15 ofthe light chain variable region; (ii) the amino acid position 77, 78 or 79 ofthe light chain variable region; (iii) the amino acid position 11, 12, 13, 14 or 15 ofthe heavy chain variable region; (iv) the amino acid position 82B, 82C or 83 ofthe heavy chain variable region; (v) any amino acid position ofthe peptide linker; (vi) adjacent to the C-terminus of polypeptide
  • polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
  • the invention is further directed to a genetic sequence encoding a single- chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
  • the invention is further directed to a genetic sequence encoding a single- chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising: (a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
  • the genetic sequence may be DNA or RNA.
  • the invention is directed to a replicable cloning or expression vehicle comprising the above described DNA sequence.
  • the invention is also directed to such vehicle which is a plasmid.
  • the invention is further directed to a host cell transformed with the above described DNA.
  • the host cell may be a bacterial cell, a yeast cell or other fungal cell, an insect cell or a mammalian cell line.
  • a preferred host is Pichia pastor is.
  • the invention is directed to a method of producing a single-chain antigen- binding polypeptide capable of polyalkylene oxide conjugation, comprising: (a) providing a first genetic sequence encoding a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
  • the invention is further directed to a multivalent single-chain antigen- binding protein, comprising two or more single-chain antigen-binding polypeptides, each single-chain antigen-binding polypeptide comprising:
  • the single-chain antigen-binding polypeptide has at least one Cys residue wherein the Cys residue is capable of polyalkylene oxide conjugation and the Cys residue is located at a position selected from the group consisting of (i) the amino acid position 11 , 12, 13, 14 or 15 ofthe light chain variable region; (ii) the amino acid position 77, 78 or 79 ofthe light chain variable region; (iii) the amino acid position 11, 12, 13, 14 or 15 ofthe heavy chain variable region; (iv) the amino acid position 82B, 82C or 83 ofthe heavy chain variable region; (v) any amino acid position ofthe peptide linker; (vi) adjacent to the C-terminus of polypeptide (a) or (b); and (vii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
  • the Cys polyalkylene oxide conjugation sequence may be capable of attaching a polyalkylene oxide moiety and the Cys residue is located at a position selected from the group consisting of (i') the amino acid position 77 ofthe light chain variable region; (ii') the amino acid position 82B ofthe heavy chain variable region; (iii') the amino acid position 3 ofthe peptide linker; (iv') adj acent to the C-terminus of polypeptide (a) or (b); (v') N-terminus and C-terminus; and (vi') combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
  • the oligo-Lys polyalkylene oxide conjugation sequence may be capable of attaching a polyalkylene oxide moiety at the oligo-Lys residues located adjacent to the C- terminus ofthe protein, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
  • the C-terminus of the second polypeptide (b) may be the native C-terminus.
  • the C-terminus ofthe second polypeptide (b) may comprise a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe second polypeptide are sufficient for the polyalkylene oxide conjugated polypeptide to be capable of binding an antigen.
  • the C-terminus of the second polypeptide may comprise an addition of one or plurality of amino acid residue(s), such that the polyalkylene oxide conjugated polypeptide is capable of binding an antigen.
  • the first polypeptide (a) may comprise the antigen binding portion ofthe variable region of an antibody light chain and the second polypeptide (b) comprises the antigen binding portion ofthe variable region of an antibody heavy chain.
  • the invention is also directed to a method of detecting an antigen suspected of being in a sample, comprising:
  • the invention is further directed to a method of imaging the internal structure of an animal, comprising administering to the animal an effective amount of the polyalkylene oxide conjugated polypeptide or protein of the invention, wherein the polyalkylene oxide conjugated polypeptide is conjugated to one or plurality of detectable label or chelator molecule(s), or conjugated to a carrier having one or plurality of detectable label or chelator molecule(s) bound to the carrier, and measuring detectable radiation associated with the animal.
  • Animal includes human and nonhuman.
  • the invention is also directed to a method for treating a targeted disease, comprising administering an effective amount of a composition comprising the polyalkylene oxide conjugated polypeptide or protein of the invention and a pharmaceutically acceptable carrier vehicle, wherein the polyalkylene oxide conjugated polypeptide is conjugated to one or plurality of bioactive molecules, such as peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, boron addend or radioisotope molecule(s), or conjugated to a carrier having one or plurality of peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, boron addend or radioisotope molecule(s) bound to the carrier.
  • bioactive molecules such as peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, boron addend or radio
  • Figure 1 is a graphical representation of three competition ELISA's in which unlabeled PEG modified CC49/212 SCA (closed squares), CC49/212 SCA (open squares), CC49 IgG (open circles), and MOPC-21 IgG (+) competed against a CC49 IgG radiolabeled with 125 I for binding to the TAG-72 antigen on a human breast carcinoma extract.
  • Figure 2 shows the DNA and protein sequence of CC49/218 SCA which has four engineered cysteine residues at the positions indicated by the codons underlined and marked by an asterisk. Also highlighed are the CDR sequences (double underlined) and the 218 linker (underlined and labeled).
  • cysteine residues in the protein which are involved in two disulfide bonds. These are not underlined.
  • the four engineered cysteine residues occur independently in four different mutants currently, but may be combined in the exact four-mutant codon version shown in this figure.
  • Figure 3 shows the DNA and protein sequence of CC49/218 SCA with an engineered oligo-lysine C-terminal tail segment.
  • the eight new lysine residues were genetically engineered at a BstEII site and are shown underlined and marked with asterisks. Also highlighted are the CDR sequences (double underlined), the 218 linker (underlined and labeled) and selected restriction sites.
  • Figure 4 is a graphical representation of three competition ELISA's in which unlabeled SC-PEG unreacted CC49/218 SCA (closed squares), CC49/218 SCA (open squares), unlabeled XUS-PEG unreacted CC49/218 SCA (open circles), SC-PEG modified CC49/218 SCA (closed circles), XUS-PEG modified CC49/218 SCA (open triangles), CC49 IgG (closed triangles), an Anti-FITC SCA (dashed line) or BL-3 IgG (dotted line) were competed against a CC49 IgG radiolabeled with 125 I for binding to the TAG- 72 antigen on a human breast carcinoma extract.
  • Figure 5 shows the pharmacokinetics of plasma retention of SCA and PEG-SCA.
  • the details ofthe experiment are described in Example 13.
  • Figure 6 shows an SDS-PAGE of the purified CC49-multimers cross- linked by PEG5000 under reducing conditions. The details ofthe experiment are described in Example 14.
  • the lanes ofthe gel contain the following: 1) trimeric form; 2) dimeric form; 3) dimeric form; 4) mixed population; 5) native CC49; 6) PEG-CC49 monomer; 7) PEG-CC49 monomer; 8) empty; 9) empty; and 10) molecular weight standards.
  • Figure 7 shows the binding kinetics of Mono-, Di-, Tri-, -PEG-CC49. The details of the experiment are described in Example 14.
  • Native CC49 is represented by the solid box.
  • PEG-mono-CC49 is represented by the open box.
  • PEG-Di-CC49 is represented by the solid diamond.
  • PEG-Tri-CC49 is represented by the open diamond.
  • Figure 8 shows the results ofthe competition assay performed in Example 16.
  • Nat is native CC49-SCA
  • C2 is PEG SC2000-CC49-SCA
  • GC is glyco- CC49-SCA
  • B* is the biotinylated CC49-SCA
  • C 12 is the PEG-SC 12,000-CC49- SCA
  • F5 is PEG-Flan-5000-CC49-SCA
  • C20 is PEG-SC20000-CC49-SCA.
  • the present invention is directed to the novel combination of a polyalkylene glycol and a single chain polypeptide having binding affinity for an antigen, the polyalkylene glycol and polypeptide preferably being joined together by means of a coupling agent.
  • Single Chain Polypeptides Single Chain Polypeptides
  • the invention relates to the discovery that polyalkylene oxide conjugated single-chain antigen-binding proteins ("SCA") or single-chain variable fragments of antibodies (“sFv”), such as PEGylated SCA proteins, have significant utility beyond that of the nonPEGylated single-chain antigen-binding proteins.
  • SCA polyalkylene oxide conjugated single-chain antigen-binding proteins
  • sFv single-chain variable fragments of antibodies
  • PEGylated SCA protein has significant utility beyond that of the nonPEGylated single-chain antigen-binding proteins.
  • PEGylated SCA protein has a PEG moiety which reduces antigenicity and increases the halflife ofthe modified polypeptide in the bloodstream.
  • the invention is directed to monovalent and multivalent SCA proteins capable of PEGylation, compositions of monovalent and multivalent PEGylated SCA proteins, methods of making and purifying monovalent and multivalent PEGylated SCA proteins, and uses for PEGylated SCA proteins.
  • the invention is also directed to PEGylated SCA proteins having a diagnostic or therapeutic agent covalently attached to an Cys- linked PEGylated polypeptide or an oligo-Lys linked PEGylated polypeptide.
  • SCA single-chain antigen-binding molecule
  • SCA single-chain antigen-binding molecule
  • Fv Fv
  • sFv Fv
  • V L variable region of an antibody
  • N H variable region of an antibody
  • the single polypeptide chain thus comprises a pair of variable regions connected by a polypeptide linker.
  • the regions may associate to form a functional antigen- binding site, as in the case wherein the regions comprise a light-chain and a heavy- chain variable region pair with appropriately paired complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • the single-chain protein is referred to as a "single- chain antigen-binding protein" or "single-chain antigen-binding molecule.”
  • Single-chain Fvs can and have been constructed in several ways. Either V L is the N-terminal domain followed by the linker and V H (a V L -Linker- V H construction) or V H is the N-terminal domain followed by the linker and V L (V H -Linker- V L construction).
  • the preferred embodiment contains V L in the N- terminal domain (see, Anand, N.N., et al, J. Biol Chem. 266:21874-21879 (1991)).
  • multiple linkers have also been used.
  • SCA sFv
  • the Fv domains have been selected from the group of monoclonal antibodies known by their abbreviations in the literature as 26-10, MOPC 315, 741F8, 520C9, McPC 603, D1.3, murine phOx, human phOx, RFL3.8 sTCR, lA6, Sel55-4, 18-2-3, 4-4-20, 7A4-1, B6.2, CC49, 3C2, 2c, MA- 15C5/K 12 G 0 , Ox, etc. ⁇ see, Huston, J.S. et al, Proc. Natl. Acad. Sci. USA 55:5879-5883 (1988); Huston, J.S.
  • Linkers of the invention used to construct sFv (SCA) polypeptides are designed to span the C-terminus of V L (or neighboring site thereof) and the N- terminus of V H (or neighboring site thereof) or between the C-terminus of V H and the N-terminus of V L .
  • the preferred length ofthe peptide linker should be from 2 to about 50 amino acids. In each particular case, the preferred length will depend upon the nature ofthe polypeptides to be linked and the desired activity ofthe linked fusion polypeptide resulting from the linkage. Generally, the linker should be long enough to allow the resulting linked fusion polypeptide to properly fold into a conformation providing the desired biological activity.
  • the appropriate linker length may be estimated by consideration ofthe 3 -dimensional conformation ofthe substituent polypeptides and the desired conformation ofthe resulting linked fusion polypeptide. Where such information is not available, the appropriate linker length may be empirically determined by testing a series of linked fusion polypeptides with linkers of varying lengths for the desired biological activity. Such linkers are described in detail in WO 94/12520, inco ⁇ orated herein by reference.
  • Preferred linkers used to construct sFv (SCA) polypeptides have between 10 and 30 amino acid residues.
  • the linkers are designed to be flexible, and it is recommended that an underlying sequence of alternating Gly and Ser residues be used.
  • three charged residues may be included, two positively charged lysine residues (K) and one negatively charged glutamic acid residue (E).
  • K positively charged lysine residues
  • E negatively charged glutamic acid residue
  • one ofthe lysine residues is placed close to the N-terminus of V H , to replace the positive charge lost when forming the peptide bond ofthe linker and the V H .Such linkers are described in detail in U.S. Patent Application S. N.
  • a n object of the present invention is to produce a single-chain antigen- binding polypeptide-polyalkylene oxide conjugate which retains antigen binding affinity within a range of about two-fold to about ten-fold ofthe antigen binding affinity ofthe native single-chain antigen-binding polypeptide.
  • a further object ofthe present invention is to produce an sFv (SCA) having three or more consecutive Lys residues such that the Lys residues are capable of being conjugated with PEG and the PEGylated polypeptide is capable of binding an antigen (i.e., the PEGylated polypeptide's ability to bind an antigen is not disrupted).
  • SCA novel sFv
  • PEG polyethylene glycol
  • the activated PEG molecules would be thiol-reactive or amine-reactive polymers such as are well known in the art.
  • the designed changes correspond to amino acid residues on the sFv (SCA) surface which are well separated spatially from the antigen-binding site as deduced from known three-dimentional models ofthe antibody Fv domain.
  • a further object ofthe invention is to produce monovalent and multivalent sFvs (SCA) having one or more Cys PEG conjugation sequence(s).
  • a further object of the invention is to produce monovalent and multivalent sFvs (SCA) having three or more consecutive Lys (i.e., oligo-Lys) PEG conjugation sequence(s).
  • SCA monovalent and multivalent sFv
  • SC As the association of two or more sFvs
  • SC As may be generated by chemically crosslinking two sFvs (SCAs) with C-terminal cysteine residues (Cumber et al , J. Immunol.
  • Multivalent antigen-binding fusion proteins of the invention can be made by any process, but preferably according to the process formaking multivalent antigen-binding proteins set forth in WO 93/11161, inco ⁇ orated herein by reference.
  • Cys PEGylation sites may occur in the V L and V H regions, adjacent to the C-terminus ofthe polypeptide (V L , V H or neighboring site thereof), the N-terminus ofthe polypeptide (V L , V H or neighboring site thereof), the linker region between the first and second polypeptide regions, or occur in a combination of these regions.
  • oligo-Lys PEGylation sites may occur in the polypeptide linker or in the C-terminus or adjacent to the C-terminus of the polypeptide.
  • the Cys or the oligo-Lys PEGylation site may occur in (1) the native C- terminus of V L (or V H ), (2) the C-terminus of V L (or V H ) wherein the C-terminus has a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe peptide are sufficient for the PEGylated polypeptide to be capable of binding an antigen or (3) the C-terminus of V L (or V H ) wherein the C-terminus has an addition of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe peptide are sufficient for the PEGylated polypeptide to be capable of binding an antigen.
  • C-terminus By “native” is intended the naturally occurring C-terminus ofthe immunoglobulin (first or second polypeptide).
  • C-terminus it is well understood in the art as intending the C-terminal amino acid residue or the C-terminal region of the polypeptide, which could include up to all of the amino acid residues of the polypeptide excluding the first N-terminal amino acid residue ofthe polypeptide.
  • C-terminus is intended as the C-terminal amino acid residue ofthe above mentioned three types of C-terminus (1 , 2, or 3), unless otherwise indicated or intended.
  • PEGylation sites were identified and engineered at residues within loop sites in regions of the sFv (SCA) that are diametrically opposed to the antigen binding site.
  • SCA sFv
  • the five loop regions and C-terminal extension chosen as preferred sites of glycosylation are among the most distant regions spatially removed from the binding site.
  • SCA sFv
  • the nucleic and amino acid sequences of each loop are examined for possible Cys PEGylation sites that may be engineered into the loop region.
  • the engineered placement ofthe Cys residue anywhere in these six identified regions can generate a preferred site for sFv (SCA) PEGylation.
  • the engineered placement of the oligo-Lys residues in the linker, the C-terminus ofthe sFv (SCA)andVor adjacent to the C-terminus of the sFv (SCA) can generate a preferred site for sFv PEGylation.
  • Figure 2 shows the following resulting designs: designed PEGylation site no. 1 in the light chain ofthe CC49/218 SCA; designed PEGylation site no. 2 in the N-terminal end ofthe linker in CC49/218 SCA; designed PEGylation site no. 3 in the heavy chain ofthe CC49/218 SCA; designed PEGylation site no. 4 at the C-terminus of the CC49/218 SCA.
  • Figure 3 shows the following resulting designs: designed oligo-Lys "hot spot" PEGylation sites at the C-terminus ofthe CC49/218 SCA. Any combination of these sites could be used.
  • nucleotide sequence which is used to introduce a Cys or oligo-Lys PEGylation site into the various positions will depend upon the naturally-occurring nucleotide sequence.
  • the most preferred sites are those in which it takes a minimum number of changes to generate the PEGylation site.
  • a particular amino acid may be encoded by multiple nucleotide sequences.
  • Site-directed mutagenesis is used to change the native protein sequence to one that inco ⁇ orates the Cys residue or oligo-Lys residues for PEGylation.
  • the mutant protein gene is placed in an expression system, such as bacterial cells, yeast or other fungal cells, insect cells or mammalian cells.
  • the mutant protein can be purified by standard purification methods.
  • Oligonucleotide-directed mutagenesis methods for generating the Cys or oligo-Lys PEGylation sites and related techniques for mutagenesis of cloned DNA are well known in the art. See, Sambrook et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1989); Ausubel et al. (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley and Sons (1987), both inco ⁇ orated herein by reference.
  • a preferred oligonucleotide-directed mutagenesis method for the present invention is according to Ho et al, Gene 77:51 -59 (1989), inco ⁇ orated herein by reference. Hosts and Vectors
  • the mutated DNA can be inserted into a cloning vector for further analysis, such as for confirmation ofthe DNA sequence.
  • the DNA sequence is operably linked to regulatory sequences controlling transcriptional expression and introduced into either a prokaryotic or eukaryotic host cell.
  • sFvs are typically produced by prokaryotic host cells
  • eukaryotic host cells are the preferred host cells.
  • Preferred host cells include yeast or other fungal cells, insect cells or mammalian cells. Standard protein purification methods may be used to purify these mutant proteins. Only minor modification to the native protein's purification scheme may be required.
  • DNA molecules such as purified genetic sequences or plasmids or vectors encoding the sFv (SCA) ofthe invention that have engineered Cys residues and/or oligo-Lys residues capable of PEG conjugation.
  • the DNA sequence for the PEGylated sFv (SCA) polypeptide can be chosen so as to optimize production in organisms such as prokaryotes, yeast or other fungal cells, insect cells or mammalian cells.
  • the DNA molecule encoding an sFv (SCA) having Cys residues and/or oligo-Lys residues for PEG conj ugation can be operably linked into an expression vector and introduced into a host cell to enable the expression ofthe engineered sFv (SCA) protein by that cell.
  • a DNA sequence encoding an sFv (SCA) having Cys and/or oligo-Lys PEGylation sites may be recombined with vector DNA in accordance with conventional techniques. Recombina- nt hosts as well as methods of using them to produce single chain proteins ofthe invention are also provided herein.
  • sFv (SCA) proteins of the invention can be accomplished in procaryotic cells.
  • Preferred prokaryotic hosts include, but are not limited to, bacteria such as Neisseria, Mycobacte ia, Streptococci, Chlamydia and E. coli.
  • Eukaryotic hosts for cloning and expression of such sFv (SCA) proteins ofthe invention include insect cells, yeast, fungi, and mammalian cells (such as, for example, human or primate cells) either in vivo, or in tissue culture.
  • a preferred host for the invention is Pichia pastor is.
  • the sFv (SCA) encoding sequence having Cys residues and/or oligo-Lys residues for PEG conjugation and an operably linked promoter may be introduced into a recipient prokaryotic or eukaryotic cell either as a non-replicating DNA (or RNA) molecule, which may either be a linear molecule or, more preferably, a closed covalent circular molecule. Since such molecules are incapable of autonomous replication, the expression of the desired sFv (SCA) protein may occur through the transient expression ofthe introduced sequence. Alternatively, permanent expression may occur through the integration of the introduced sFv (SCA) sequence into the host chromosome.
  • the sFv (SCA) sequence can be integrated into the host cell chromosome.
  • Cells which have stably integrated the introduced DNA into their chromosomes can be selected by also introducing one or more markers which allow for selection of host cells which contain the sFv (SCA) sequence and marker.
  • the marker may complement an auxotrophy in the host (such as his4, leu2, or ura3, which are common yeast auxotrophic markers), biocide resistance, e.g., antibiotics, or resistance to heavy metals, such as copper, or the like.
  • the selectable marker gene can either be directly linked to the sFv (SCA) DNA sequence to be expressed, or introduced into the same cell by co-transfection.
  • the introduced sequence will be inco ⁇ orated into a plasmid vector capable of autonomous replication in the recipient host cell.
  • a plasmid vector capable of autonomous replication in the recipient host cell.
  • Any of a wide variety of vectors may be employed for this pu ⁇ ose. Factors of importance in selecting a particular plasmid or viral vector include: the ease with which recipient cells that contain the vector may be recognized and selected from those recipient cells which do not contain the vector; the number of copies ofthe vector which are desired in a particular host; and whether it is desirable to be able to "shuttle" the vector between host cells of different species.
  • yeast vector systems Any of a series of yeast vector systems can be utilized. Examples of such expression vectors include the yeast 2-micron circle, the expression plasmids YEP 13, YCP and YRP, etc., or their derivatives. Such plasmids are well known in the art (Botstein et al, Miami Wntr. Symp. 19:265-274 (1982); Broach, J.R., In: The Molecular Biology of the Yeast Saccharomyces: Life Cycle and Inheritance, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, p. 445- 470 (1981); Broach, J.R., Cell 25:203-204 (1982)).
  • vectors For a mammalian host, several possible vector systems are available for expression.
  • One class of vectors utilize DNA elements which provide autonomously replicating extra-chromosomal plasmids, derived from animal viruses such as bovine papilloma viras, polyoma virus, adenovirus, or SV40 virus.
  • a second class of vectors relies upon the integration ofthe desired gene sequences into the host chromosome.
  • Cells which have stably integrated the introduced DNA into their chromosomes may be selected by also introducing one or more markers which allow selection of host cells which contain the expression vector.
  • the marker may provide for prototrophy to an auxotrophic host, biocide resistance, e.g., antibiotics, or resistance to heavy metals, such as copper or the like.
  • the selectable marker gene can either be directly linked to the DNA sequences to be expressed, or introduced into the same cell by co-transformation. Additional elements may also be needed for optimal synthesis of mRNA. These elements may include splice signals, as well as transcription promoters, enhancers, and termination signals.
  • the cDNA expression vectors inco ⁇ orating such elements include those described by Okayama, H., Mol. Cell. Biol. 3:280 (1983), .and others.
  • vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNHl ⁇ a, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia.
  • preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
  • Preferred vectors for expression in Pichia are pHIL-S 1 (Invitrogen Co ⁇ .) and pPIC9 (Invitrogen Co ⁇ .). Other suitable vectors will be readily apparent to the skilled artisan.
  • the DNA constructs may be introduced or transformed into an appropriate host.
  • Various techniques may be employed, such as transformation, transfection, protoplast fusion, calcium phosphate precipitation, electroporation, or other conventional techniques.
  • the cells After the cells have been transformed with the recombinant DNA (or RNA) molecule, the cells are grown in media and screened for appropriate activities. Expression of the sequence results in the production of the mutant sFv (SCA) for PEG conjugation of the present invention.
  • SCA mutant sFv
  • R is selected from the group consisting of hydrogen, lower alkyl, and mixtures thereof
  • R 1 is selected from the group consisting of hydrogen and lower alkyl
  • n is a positive integer.
  • lower alkyl is meant an alkyl group having from one to four carbon atoms, i.e., methyl, ethyl, propyl, butyl, and isomers of the foregoing.
  • R is preferably selected from the group consisting of hydrogen, methyl, and mixtures thereof
  • R 1 is preferably selected from the group consisting of hydrogen and methyl
  • n is preferably a positive integer of 500 or less.
  • R is most preferably hydrogen
  • R 1 is most preferably methyl
  • n is most preferably an integer of 7 to 150.
  • poly(alkylene glycols) employed in the practice ofthe present invention are poly(ethylene glycol), poly(propylene glycol), mixtures thereof, and copolymers of poly(ethylene glycol) and poly(propylene glycol), wherein one of the terminal hydroxyl groups ofthe polymer may be substituted with a lower alkyl group.
  • a preferred polyalkylene glycol for use in the present invention is poly(ethylene glycol)-hydrazine.
  • the most preferred polyalkylene glycol for use in the present invention is methoxy poly(ethylene glycol).
  • the polyalkylene glycol employed in the practice ofthe present invention will be designated PAG, which term is intended to include both compounds wherein R 1 is hydrogen and compounds wherein R 1 is alkyl.
  • PEG refers to poly(ethylene glycol) and mPEG refers to methoxy poly(ethylene glycol).
  • the PAG does not have to be of a particular molecular weight, but it is preferred that the molecular weight be between about 500 and about 40,000; more preferably, between about 2,000 and about 20,000.
  • the choice of molecular weight of PAG is made based on the nature of the particular polypeptide employed, for example, the number of amino or other groups available on the polypeptide for modification. Molecular weights of about 10,000 and about 20,000 are most preferred.
  • PAGs that contain two terminal hydroxyl groups per moiety are capable of crosslinking other polymers, e.g. proteins. Where, as is often the case, crosslinking would be deemed undesirable, such crosslinking can be minimized or prevented by means known in the art.
  • a preferred means for preventing crosslinking is to preblock one end ofthe PAG, such as is done in the commercially available methoxy poly(ethylene glycol).
  • the P AGs employed in the practice ofthe present invention are preferably coupled to polypeptides by means of suitable coupling agents.
  • suitable coupling agents A useful review of a number of coupling agents that can be employed in the practice ofthe present invention appears in Dreborg etal, Critical Reviews in Therapeutic Drug Carrier
  • the terminal hydroxyl groups of PEG can be transformed into amine, carboxyl, or hexamethyl isocyanate groups. See, for example, Zalipsky et al, 1983, supra.
  • a mixed anhydride derivative of carboxylated mPEG can be prepared in the presence of triethylamine and then reacted with proteins:
  • Carboxylated mPEG can also be reacted with hydroxysuccinimide in the presence of dicyclohexylcarbodumide and dimethyl formamide for reaction with protein:
  • Veronese et al, Appl. Biochem. & Biotechnol 77:141-152 (1985) describe the activation of methoxy poly(ethylene glycol) with phenylchloroformates, e.g., 2,4,5-trichlorophenylchloroformate or / nitrophenylchloroformate. These derivatives are linked to peptides by urethane linkages:
  • Zalipsky in U.S. Patent No. 5,122,614, describes the activation of PEG by conversion into its N-succinimide carbonate derivative (“SC-PEG”): o o
  • BSC-PEG polyethylene glycol-bis-succinidyl carbonate
  • activated PAGs can also be employed in the practice ofthe present invention.
  • the preferred activated PAG for use in the practice ofthe present invention is selected from the group consisting of SS-PEG and SC-PEG.
  • SC-PEG is most preferred.
  • the invention further provides for the use of branched, substantially non- antigenic polymers for polyalkylene oxide conjugation ofthe sFv (SCA) proteins corresponding to the formula:
  • (A) represents an activated functional group capable of undergoing nucleophilic substitution.
  • (A) can be a group which is capable of bonding with biologically active nucleophiles or moieties capable of doing the same.
  • R includes a poly(alkylene oxide) PAO such as poly(ethylene glycol) PEG or mPEG. It is preferred that each chain have a molecular weight of between about 200 and about 12,000 daltons and preferably between about 1,000 and about 10,000 daltons. Molecular weights of about 5,000 daltons are most preferred.
  • PAO poly(alkylene oxide) PAO
  • PAO poly(ethylene glycol) PEG or mPEG. It is preferred that each chain have a molecular weight of between about 200 and about 12,000 daltons and preferably between about 1,000 and about 10,000 daltons. Molecular weights of about 5,000 daltons are most preferred.
  • 2 or 3 polymer chains are joined to the alphatic linking moiety (L).
  • Suitable aliphatics included substituted alkyl diamines and triamines, lysine esters and malonic ester derivatives.
  • the linking moieties are perferably non-planar, so that the polymer chains are not rigidly fixed.
  • the linking moiety (L) is also a means for attaching the mulitple polymer chains or "branches" to (A), the moeity through which the polymer attaches to the sFv (SCA) protein.
  • (L) preferably includes a multiply-functionalized alykyl group containing up to 18, and more preferably between 1-10 carbon atoms.
  • a heteroatom such as nitrogen, oxygen or sulfur may be included within the alkyl chain.
  • the alkyl chain may also be branched at a carbon or nitrogen atom.
  • (L) is a single nitrogen atom.
  • (L) and (R) are preferably joined by a reaction between nucleophilic functional groups on both (R) and (L). Each (R) is suitably functionalized to undergo nucleophilic substitution and bond with (L). Such functionalization of polymers is readily apparent to those of ordinary skill in the art. A wide variety of linkages are contemplated between (R) and (L).
  • Urethane (carbamate) linkages are preferred.
  • the bond can be formed, for example, by reacting an amino group such as l,3-diamino-2-propanol with methoxypolyethylene glycol succinimidyl carbonate as described in U.S. Patent No. 5,122,614.
  • -Amide linkages which can be formed by reacting an amino- terminated non-antigenic polymer suchas methoxypolyethylene glycol-amine (mPEG amine) with an acyl chloride functional group. Examples of other such linkages include ether, amine, urea, and thio and thiol analogs thereof, as well as the thio and thiol analogs ofthe urethane and amide linkages discussed supra.
  • the moiety (A) of Formula II represents groups that "activate" the branched polymers ofthe present invention for conjugation with biologically active materials.
  • (A) can be a moiety selected from:
  • Functional groups capable of reacting with an amino group such as: a) carbonates such as the p-nitrophenyl or succinimidyl; b) carbonyl imidazole; c) azlactones; d) cyclic imide thiones; or e) isocyanates or isothiocyanates.
  • Functional groups capable of reacting with carboxylic acid groups and reactive with carbonyl groups such as: a) primary amines; or b) hydrazine and hydrazide functional groups such as the acyl hydrazides, carbazates, semicarbamates, thiocarbazates, etc.
  • Functional groups capable of reacting with mercapto or sulfhydryl groups such as phenyl glyoxals; see, for example, U.S. Patent No. 5,093,531.
  • the moiety (A) can also include a spacer moiety located proximal to the aliphatic linking moiety (L).
  • the spacer moiety may be a heteroalkyl, alkoxyl, alkyl containing up to 18 carbon atoms or even an additional polymer chain.
  • the spacer moieties can be added using standard synthesis techniques.
  • the branched polymers generally, U-PAO's or U-PEG's, are formed using conventional reaction techniques known to those of ordinary skill in the art.
  • umbrella-like branched polymers ofthe present invention react with biologically active nucleophiles to form conjugates.
  • the point of polymer attachment depends upon the functional group (A). For example,
  • (A) can be a succinimidyl succinate or carbonate and react with e-amino lysines.
  • the branched polymers can also be activated to link with any primary or secondary amino group, mercapto group, carboxylic acid group, reactive carbonyl group or the like found on biologically active polypeptides. Other groups are apparent to those of ordinary skill in the art.
  • branched polymers One ofthe main advantages ofthe use ofthe branched polymers is that the branching imparts an umbrella-like three dimensional protective covering to the materials they are conjugated with. This contrasts with the string-like structure ofthe straight chain polymers discussed, supra. .An additional advantage of the branched ploymers is that they provide the benefits associated with attaching several strands of polymers to a sFv protein but require substantially fewer conjugation sites. The desired properties of PEGylation are realized and the loss of bioactivity is minimized.
  • One or more of the activated branched polymers can be attached to a biologically active nucleophile, such as an sFv protein, by standard chemical reactions.
  • a biologically active nucleophile such as an sFv protein
  • the upper limit for (z) will be determined by the number of available nucleophilic attachment sites and the degree of polymer attachment sought by the artisan.
  • the degree of conjugation can be modified by varying the reaction stoichimetry using well-known techniques. More than one polymer conjugated to the nucleophile can be obtained by reacting a stoichimetric excess ofthe activated polymer with the nucleophile.
  • the fermentation of the sFv-producing E. coli strains are performed at 32 °C using a casein digest-glucose-salts medium. At an optical density of 18 to 20 at 600 nm, sFv expression is induced by a 42° C temperature shock for one hour. After the fermentation is cooled to 10°C, the cells are harvested by centrifugation at 7000g for ten minutes. The wet cell paste is then stored frozen at -20 °C. Approximately 200 to 300 g of wet cell paste is normally recovered from one 10-liter fermentation.
  • the cell paste from three 10-liter fermentations (600- 900 g) is thawed overnight at 4 °C and gently resuspended at 4 °C in 50 mM Tris- HC1, 1.0 mM EDTA, 100 mM KC1, 0J mM phenylmethylsulfonyl chloride (PMSF), pH 8.0 (lysis buffer), using 10 liters of lysis buffer for every kilogram of wet cell paste.
  • PMSF mM phenylmethylsulfonyl chloride
  • pH 8.0 pH 8.0
  • the chilled mixture is passed three times through a Manton-Gaulin cell homogenizer to fully lyse the cells.
  • the cell homogenizer raises the temperature ofthe cell lysate to 25 ⁇ 5 °C
  • the cell lysate is cooled to 5 ⁇ 2°C with a Lauda/Brinkman chilling coil after each pass. Complete lysis is verified by visual inspection under a microscope.
  • the cell lysate is centrifuged at 24,3 OOg for thirty minutes at 6°C using a Sorvall RC-5B centrifuge.
  • the pellet contains the insoluble sFv and the supernatant is discarded.
  • the pellet is washed by gently scraping it from the centrifuge bottles and resuspending it in 5 liters of lysis buffer/kg of wet cell paste.
  • the resulting 3.0-4.5-liter suspension is again centrifuged at 24,300g for 30 min at 6°C, and the supernatant is discarded.
  • This washing ofthe cell pellet removes soluble E. coli proteins and can be repeated as many as five times. At any time during this washing procedure the material can be stored as a frozen pellet at -20°C.
  • a substantial time saving in the washing steps can be accomplished by utilizing a Pellicon tangential flow apparatus equipped with 0.22- ⁇ m microporous filters.
  • the washed cell pellet is solubilized at 4°C in freshly prepared 6 M guanidine hydrochloride, 50 mM Tris-HCl, 10 mM CaCl 2 , 50 mM KC1, pH 8.0 (denaturing buffer), using 6 ml/g of pellet. If necessary, a few quick pulses from a Heat Systems Ultrasonics tissue homogenizer can be used to complete the solubilization.
  • the resulting suspension is centrifuged at 24,300g for 45 minutes at 6°C and the pellet is discarded.
  • the optical density of the supernatant is determined at 280 nm and if the OD 2g0 is above 30, additional denaturing buffer is added to obtain an OD 280 of approximately 25.
  • the supernatant is slowly diluted into cold (4-7 °C) refolding buffer (50 mM Tris-HCl, 10 mM CaCl 2 , 50 mM KC1, 0J mM PMSF, pH 8.0) until a 1 :10 to 1 :100 dilution is reached (final volume 70-120 liters).
  • the refolding buffer should be prepared at least one day prior to use, to allow sufficient time for it to cool to 4°C. The best results will be obtained when the supernatant is slowly added to the refolding buffer over a two hour period, with gentle mixing.
  • the solution is left undisturbed for at least twenty hours and then filtered through a Millipore Pellicon tangential flow apparatus at 4°C with four to six 0.45- ⁇ m microporous membranes (HVLP 000 C5).
  • the filtrate is concentrated to 1 to 2 liters using a Pellicon apparatus with four to six 10,000 NMWL cassettes (SK1PA156A4), again at 4°C.
  • the concentrated crude sFv sample is buffer exchanged at 4°C into 20 mM 2-[N-mo ⁇ holino]ethanesulfonic acid (Mes), 0.3 mM CaCl 2 , pH 6.0, using the Pellicon ultrafiltration apparatus equipped with four to six 10,000 NMWL cassettes.
  • the sample is then chromatographed on a Waters Accell Plus CM ion- exchange (RCM) column (4.7 x 30.0 cm).
  • RCM Waters Accell Plus CM ion- exchange
  • the material Prior to loading on the HPLC, the material is filtered through a 0.22- ⁇ m filter and the Accell column is equilibrated with Buffer A (40 mM Mes, 1 mM CaCl 2 , pH 6.0). Following sample loading, the Accell column is eluted over a 55 -minute period with a linear gradient of Buffer A and Buffer B (40 mM Mes, 100 mM CaCl 2 , pH 7.0). (See
  • the Accell Plus CM column has a capacity of about 3 g and thus all the crude sFv sample can normally be loaded in a single run.
  • the fractions are analyzed using 4-20% Novex SDS-PAGE gels and the peak fractions are pooled.
  • sFv proteins have an extinction coefficient of about 2.0 mg ml "1 cm "1 at 280 nm and this can be used to determine protein concentration.
  • the sFv sample is eluted from the PolyCAT A column with a 50-min linear gradient of Buffer D and Buffer E (40 mM Mops, 10 mM Ca acetate, pH 8.0). See Table 2.
  • the sFv proteins will often elute between 20 and 26 min when this gradient is used. This corresponds to an eluting solvent composition of approximately 70% Buffer D and 30% Buffer E.
  • This purification procedure yields sFv proteins that are more than 95% pure as examined by SDS-PAGE and Scatchard analysis. Modifications ofthe above procedure may be dictated by the isoelectric point of the particular sFv being purified, which is often between 8.0 and 9.3.
  • the polyalkylene gly cols (PAGs) employed in the practice ofthe present invention which, as indicated above, are preferably activated by reaction with a coupler, can be reacted with any of several groups that may be present attached to the chain ofthe single chain antigen binding molecules, e.g. terminal carboxyl groups, thiol groups, phenolic hydroxyl groups, or primary amino groups located at the chain terminus or along the chain. It is preferred to react activated PAGs with primary amine groups, especially those occurring along the peptide chain. It is most preferred that the activated PAGs be coupled to the e amino groups of lysine residues as well as cysteine residues in the polypeptide.
  • PAGs polyalkylene gly cols
  • the reaction between the PAG and the single chain polypeptide is normally carried out in solution, preferably an aqueous buffer solution providing a pH in the range of from about 6 to about 10, preferably from about 7 to about 9, most preferably from about 7 to about 8.
  • aqueous buffer solution providing a pH in the range of from about 6 to about 10, preferably from about 7 to about 9, most preferably from about 7 to about 8.
  • the reaction between the PAG and the single chain polypeptide will normally be run under conditions that will not give rise to denaturation, e.g. mild temperatures and no more agitation than necessary.
  • the reaction will preferably be run at a temperature in the range of from about 4° C to about 25° C. More preferably, the reaction will be run at room temperature, i.e. from about 20° C to about 25° C.
  • the amount of PAG employed relative to the amount of single chain polypeptide will be dependent upon the desired nature ofthe reaction product. Where, for example, it is desired to react a PAG with each lysine residue along the polypeptide chain, an amount of PAG at least equimolar to the lysine concentration will be required. It will be advantageous to employ an excess of PAG, where possible, in order to increase the reaction rate and the likelihood of a complete reaction. Clearly, if fewer than all ofthe possible reaction sites along the polypeptide chain are to be derivatized, correspondingly less PAG will be used.
  • molar excesses of PAG's it has been determined that molar excesses on the order of 2 - 100 of the PAG can be used; molar excesses of 2 - 10 are preferred.
  • the time required for the reaction will depend upon a number of factors, such as reaction temperature, concentration of reactants, and whether full or partial reaction is desired.
  • the course of the reaction can be monitored by conventional means, such as the analysis of periodic samples by size exclusion chromatography or gel electrophoresis.
  • the reaction can conveniently be terminated when desired by the addition of a compound having a primary amine group, e.g. glycine, to scavenge the excess PAG.
  • reaction time of about 15 - 120 minutes will typically be required to fully react the PAG with the primary amine groups of the lysine residues of the single chain polypeptide at room temperature.
  • the skilled practitioner will understand that the time for conjugation, as well as the amount and type of PAG, must not be such as to inactivate the polypeptide being employed.
  • Purification ofthe PAG/single chain polypeptide reaction product can be effected by means commonly employed by those skilled in the art, such as, for example, size exclusion chromatography, ion-exchange chromatography, ultrafiltration, dialysis, and the like. Solutions of the reaction product can, if desired, be concentrated with a rotary evaporator and can be obtained in the dry state by lyophilization.
  • the resulting adduct is expected to be useful both diagnostically and therapeutically, exhibiting, as compared to the unreacted single chain polypeptide, decreased immunogenicity, increased circulating life, and increased stability while maintaining an acceptable level of activity.
  • the single chain antigen binding polypeptide can be reacted with the activated branched polyethylene glycol polymers discussed above in an aqueous reaction medium which can be buffered, depending on the pH requirements ofthe nucleophile.
  • the optimum pH for the reaction is generally between about 6.5 and about 8.0 and preferably about 7.4 for polypeptides.
  • the optimum reaction conditions for the sFv stability, reaction efficiency, etc. is within the level of ordinary skill in the art.
  • the preferred temperature range is between 4°C and 37 °C. The reaction temperature cannot exceed the temperature at which the nucleophile may denature or decompose. It is preferred that the nucleophile be reacted with an excess ofthe activated branched polymer.
  • the cojugate is recovered and purified, for example, by diafiltration, column chromatography, combinations thereof, or the like.
  • the polyalkylene oxide conjugated sFv may further be modified by conjugating a diagnostic or therapeutic agent to the polyalkylene oxide conjugated sFv.
  • a diagnostic or therapeutic agent to the polyalkylene oxide conjugated sFv.
  • the general method of preparing an antibody conjugate according to the invention is described in Shih, L.B., etal, Cancer Res. 57:4192 (1991); Shih, L.B., andD.M. Goldenberg, Cancer Immunol. Immunother. 31:197 (1990); Shih, L.B., et al, Intl. J. Cancer 46:1101 (1990); Shih, L.B., et al, Intl. J.
  • the indirect method involves reacting an antibody (or sFv), whose polyalkylene oxide has a functional group, with a carrier polymer loaded with one or plurality of bioactive molecules, such as, peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, chelator, boron addend or detectable label molecule(s).
  • a carrier polymer loaded with one or plurality of bioactive molecules, such as, peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, chelator, boron addend or detectable label molecule(s).
  • the polyalkylene oxide conjugated sFv may be directly conjugated with a diagnostic or therapeutic agent.
  • the general procedure is analogous to the indirect method of conjugation except that a diagnostic or therapeutic agent is directly attached to an oxidized sFv component. See Hansen et al, U.S. Patent
  • the polyalkylene oxide conjugated sFv can be attached to a derivative of the particular drug, toxin, chelator, boron addend or label to be loaded, in an activated form, preferably a carboxyl-activated derivative, prepared by conventional means, e.g., using dicyclohexylcarbodumide (DCC) or a water soluble variant thereof, to form an intermediate adduct.
  • DCC dicyclohexylcarbodumide
  • drugs and toxins are .known which have a cytotoxic effect on tumor cells or microorganisms that may infect a human and cause a lesion, in addition to the specific illustrations given above. They are to be found in compendia of drugs and toxins, such as the Merck Index and the like. . Any such drug can be loaded onto a carrier or directly onto a polyalkylene oxide conjugated sFv by conventional means well known in the art, and illustrated by analogy to those described above.
  • Chelators for radiometals or magnetic resonance enhancers are also well known in the art. Typical are derivatives of ethylenediaminetetraacetic acid (EDTA) and diethylenetriaminepentaacetic acid (DTP A). These typically have groups on the side chain by which the chelator can be attached to a carrier or directly onto a polyalkylene oxide conjugated sFv. Such groups include, e.g., a benzylisothiocyanate, by which the DTP A or EDTA can be coupled to the reactive group of an sFv.
  • EDTA ethylenediaminetetraacetic acid
  • DTP A diethylenetriaminepentaacetic acid
  • Labels such as radioisotopes, enzymes, fluorescent compounds, electron transfer agents, .and the like can also be linked to carrier or directly onto a polyalkylene oxide conjugated sFv by conventional methods well known to the art. These labels and the sFv conjugates prepared from them can be used for immunoassays and for immunohistology, much as the sFv conjugate prepared by direct attachment ofthe labels to the sFv.
  • the loading ofthe conjugates according to the present invention with a plurality of labels can increase the sensitivity of assays or histological procedures, where only low extent of binding ofthe sFv to target antigen is achieved.
  • Boron addends e.g., carboranes
  • carboranes when attached to single-chain antigen binding molecules and targeted to lesions, can be activated by thermal neutron irradiation and converted to radioactive atoms which decay by alpha emission to produce highly cytotoxic short-range effects.
  • Carboranes can be made with carboxyl functions on pendant side chains, as is well known in the art.
  • Loading of drugs on the carrier will depend upon the potency ofthe drug, the efficiency of sFv targeting and the efficacy ofthe conjugate once it reaches its target. In most cases, it is desirable to load at least 20, preferably 50, and often 100 or more molecules of a drug on a carrier.
  • the ability to partially or completely detoxify a drug as a conjugate according to the invention, while it is in circulation, can reduce systemic side effects ofthe drug and permit its use when systemic administration of the unconjugated drug would be unacceptable.
  • Administration of more molecules of the drug, but conjugated to the sFv on a carrier, according to the present invention permits therapy while mitigating systemic toxicity.
  • Toxins will often be less heavily loaded than drugs, but it will still be advantageous to load at least 5, preferably 10 and in some cases 20 or more molecules of toxin on a carrier and load at least one carrier chain on the sFv for targeted delivery.
  • the polyalkylene oxide conjugated sFv (SCA) polypeptide conjugates of the present invention are expected to have much longer circulating half lifes and reduced immunogenicity in vivo. This may solve a potential limitation relating to very rapid blood clearance of some sFv proteins. It would also reduce or eliminate concerns about repeated administration of a therapeutic sFv which may otherwise provoke an immune response in the patient.
  • the choice of the particular cysteine and/or oligo-lysine mutant combinations may allow one to achieve circulating lives over a considerable range depending on the specific polyalkylene oxide conjugated sFv variant polypeptide. This would allow sFv to be administered for the therapeutic use of choice.
  • a diagnostic or therapeutic agent is a molecule or atom which is conjugated to an antibody and useful for diagnosis or for therapy. The immunoreactivity ofthe antibody is retained. Diagnostic or therapeutic agents include drugs, toxins, chelators, boron compounds and detectable labels. See “Conjugates" section, supra, for further details.
  • the diagnostic or therapeutic agent may be, but is not limited to, at least one selected from a nucleic acid, a compound, a protein, an element, a lipid, an antibody, a saccharide, an isotope, a carbohydrate, an imaging agent, a lipoprotein, a glycoprotein, an enzyme, a detectable probe, or any combination thereof, which may be detectably labeled as for labeling antibodies, as described herein.
  • labels include, but are not limited to, enzymatic labels, radioisotope or radioactive compounds or elements, fluorescent compounds or metals, chemiluminescent compounds and bioluminescent compounds.
  • any other known diagnostic or therapeutic agent can be used in a method ofthe present invention.
  • a therapeutic agent used in the present invention may have a therapeutic effect on the target cell, the effect selected from, but not limited to, correcting a defective gene or protein, a drug action, a toxic effect, a growth stimulating effect, a growth inhibiting effect, a metabolic effect, a catabolic affect, an anabolic effect, an antiviral effect, an antibacterial effect, a hormonal effect, a neurohumoral effect, a cell differentiation stimulatory effect, a cell differentiation inhibitory effect, a neuromodulatory effect, an antineoplastic effect, an anti-tumor effect, an insulin stimulating or inhibiting effect, a bone marrow stimulating effect, a pluripotent stem cell stimulating effect, an immune system stimulating effect, and any other known therapeutic effects that may be provided by a therapeutic agent delivered to a cell via a delivery system according to the present invention.
  • the sFv conjugate of the present invention may be used for protection, suppression or treatment of infection or disease.
  • protection from infection or disease as used herein is intended “prevention,” “suppression” or “treatment.”
  • Prevention involves administration of a glycosylated sFv conjugate prior to the induction ofthe disease.
  • suppression involves administration ofthe composition prior to the clinical appearance ofthe disease.
  • Treatment involves administration ofthe protective composition after the appearance of the disease. It will be understood that in human and veterinary medicine, it is not always possible to distinguish between “preventing” and “suppressing” since the ultimate inductive event or events may be unknown, latent, or the patient is not ascertained until well after the occurrence of the event or events. Therefore, it is common to use the term “prophylaxis” as distinct from “treatment” to encompass both “preventing” and “suppressing” as defined herein.
  • the term “protection,” as used herein, is meant to include “prophylaxis.”
  • Such additional therapeutic agents which can further comprise a therapeutic agent or composition ofthe present invention may be selected from, but are not limited to, known and new compounds and compositions including antibiotics, steroids, cytotoxic agents, vasoactive drugs, antibodies and other therapeutic modalities.
  • antibiotics used in the treatment of bacterial shock such as gentamycin, tobramycin, nafcillin, parenteral cephalosporins, etc; adrenal corticosteroids and analogs thereof, such as methyl prednisolone, mitigate the cellular injury caused by endotoxins; vasoactive drugs, such as alpha receptor blocking agent (e.g., phenoxybenzamine), beta receptor agonists (e.g., isoproterenol), and dopamine are agents suitable for treating septic shock.
  • alpha receptor blocking agent e.g., phenoxybenzamine
  • beta receptor agonists e.g., isoproterenol
  • dopamine are agents suitable for treating septic shock.
  • Polyalkylene oxide conjugated sFv ofthe invention may also be used for diagnosis of disease and to monitor therapeutic response.
  • Other uses of polyalkylene oxide conjugated sFv proteins are specific targeting of pro-drug activating enzymes to tumor cells by a bispecific molecule with specificity for tumor cells and enzyme.
  • Polyalkylene oxide conjugated sFv may be used for specific delivery of drug to an in vivo target, such as a tumor, delivery of radioactive metals for tumor radioimmunodiagnosis or radioimmunotherapy (Goldenberg, D.M., Am. J. Med.
  • Affinity purification is made possible by affixing the polyalkylene oxide conjugated sFv protein to a support, with the antigen-binding sites exposed to and in contact with the ligand molecule to be separated, and thus purified.
  • Biosensors generate a detectable signal upon binding of a specific antigen to an antigen-binding molecule, with subsequent processing of the signal.
  • Polyalkylene oxide conjugated sFv proteins when used as the antigen-binding molecule in biosensors, may change conformation upon binding, thus generating a signal that may be detected.
  • the invention is also directed to a method of detecting an antigen suspected of being in a sample by contacting the sample with the polyalkylene oxide conjugated sFv that is labeled.
  • a sample may comprise at least one compound, mixture, surface, solution, emulsion, suspension, mixture, cell culture, fermentation culture, cell, tissue, secretion and/or derivative or extract thereof.
  • Such samples can also include, e.g., animal tissues, such as blood, lymph, cerebrospinal fluid (CNS), bone marrow, gastrointestinal contents, and portions, cells or internal and external secretions of skin, heart, lung and respiratory system, liver, spleen, kidney, pancreas, gall bladder, gastrointestinal tract, smooth, skeletal or cardiac muscle, circulatory system, reproductive organs, auditory system, the autonomic and central nervous system, and extracts or cell cultures thereof.
  • animal tissues such as blood, lymph, cerebrospinal fluid (CNS), bone marrow, gastrointestinal contents, and portions, cells or internal and external secretions of skin, heart, lung and respiratory system, liver, spleen, kidney, pancreas, gall bladder, gastrointestinal tract, smooth, skeletal or cardiac muscle, circulatory system, reproductive organs, auditory system, the autonomic and central nervous system, and extracts or cell cultures thereof.
  • CNS cerebrospinal fluid
  • Such samples can be measured using methods ofthe present invention in vitro, in
  • Such samples can also include environmental samples such as earth, air or water samples, as well as industrial or commercial samples such as compounds, mixtures, surfaces, aqueous chemical solutions, emulsions, suspensions or mixtures.
  • samples that can be used in methods ofthe present invention include cell culture and fermentation media used for growth of prokaryotic or eukaryotic cells and/or tissues, such as bacteria, yeast, mammalian cells, plant cells and insect cells.
  • prokaryotic or eukaryotic cells and/or tissues such as bacteria, yeast, mammalian cells, plant cells and insect cells.
  • eukaryotic cells and/or tissues such as bacteria, yeast, mammalian cells, plant cells and insect cells.
  • These uses include detectably-labeled forms ofthe polyalkylene oxide conjugated sFv protein.
  • Types of labels are well-known to those of ordinary skill in the art. They include radiolabeling, chemiluminescent labeling, fluorochromic labeling, and chromophoric label
  • Other uses include imaging the internal structure of an animal (including a human) by administering an effective amount of a labeled form of the polyalkylene oxide conjugated sFv protein and measuring detectable radiation associated with the animal. They also include improved immunoassays, including sandwich immunoassay, competitive immunoassay, and other immunoassays wherein the labeled antibody can be replaced by the PEGylated sFv protein of this invention. See, e.g., Kohler etal, Nature 256:495 (1975); Kohler et al, Eur. J. Immunol. 6:511 (1976); Kohler et al, Eur. J. Immunol.
  • Conjugates ofthe present invention can be formulated according to known methods to prepare pharmaceutically useful compositions, such as by admixture with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their formulation are described, for example, in Remington 's Pharmaceutical Sciences, 18th ed., Osol, A., ed., Mack, Easton PA (1990). In order to form a pharmaceutically acceptable composition suitable for effective administration, such compositions will contain a therapeutically effective amount ofthe immunoconjugate, either alone, or with a suitable amount of carrier vehicle.
  • Controlled release preparations may be achieved by the use of polymers to complex or absorb the immunoconjugate ofthe present invention.
  • the controlled delivery may be exercised by selecting appropriate macromolecules (for example, polyesters, polyamino acids, polyvinyl pyrrolidone, ethylene- vinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate).
  • the rate of drug release may also be controlled by altering the concentration of such macromolecules.
  • Another possible method for controlling the duration of action comprises inco ⁇ orating the therapeutic agents into particles of a polymeric substance such as polyesters, polyamino acids, hydrogels, poly(lactic acid) or ethylene vinylacetate copolymers.
  • microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, by the use of hydroxymethylcellulose or gelatin-microcapsules or poly(methylmethacrylate) microcapsules, respectively, or in a colloid drug delivery system, for example, liposomes, albumin microspheres, microemulsions, nanoparticles, nanocapsules, or in macroemulsions.
  • a colloid drug delivery system for example, liposomes, albumin microspheres, microemulsions, nanoparticles, nanocapsules, or in macroemulsions.
  • the immunoconjugate may be provided to a patient by means well known in the art.
  • Such means of introduction include oral means, intranasal means, subcutaneous means, intramuscular means, intravenous means, intra-arterial means, or parenteral means.
  • Intravenous, intraarterial or intrapleural administration is normally used for lung, breast, and leukemic tumors.
  • Intraperitoneal administration is advised for ovarian tumors.
  • Intrathecal administration is advised for brain tumors and leukemia.
  • Subcutaneous administration is advised for Hodgkin's disease, lymphoma and breast carcinoma.
  • Catheter perfusion is useful for metastatic lung, breast or germ cell carcinomas of the liver.
  • Intralesional administration is useful for lung and breast lesions.
  • compositions according to the invention may be administered parenterally in combination with conventional injectable liquid carriers such as sterile pyrogen-free water, sterile peroxide-free ethyl oleate, dehydrated alcohol, or propylene glycol.
  • conventional injectable liquid carriers such as sterile pyrogen-free water, sterile peroxide-free ethyl oleate, dehydrated alcohol, or propylene glycol.
  • Conventional pharmaceutical adjuvants for injection solution such as stabilizing agent, solubilizing agents and buffers, such as ethanol, complex forming agents such as ethylene diamine tetraacetic acid, tartrate and citrate buffers, and high-molecular weight polymers such as polyethylene oxide for viscosity regulation may be added.
  • Such compositions may be injected intramuscularly, intraperitoneally, or intravenously.
  • carriers and diluents include albumin and/or other plasma protein components such as low density lipoproteins, high density lipoproteins and the lipids with which these serum proteins are associated.
  • These lipids include phosphatidyl choline, phosphatidyl serine, phosphatidyl ethanolamine and neutral lipids such as triglycerides.
  • Lipid carriers also include, without limitation, tocopherol.
  • At least one polyalkylene oxide conjugated sFv linked to a therapeutic agent according to the invention may be administered by any means that achieve their intended pu ⁇ ose, for example, to treat various pathologies, such as cell inflammatory, allergy, tissue damage or other related pathologies.
  • a typical regimen for preventing, suppressing, or treating various pathologies comprises administration of an effective amount of an sFv conjugate, administered over a period of one or several days, up to and including between one week and about 24 months.
  • the dosage ofthe present invention administered in vivo or in vitro will be dependent upon the age, sex, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature ofthe effect desired.
  • the ranges of effective doses provided below are not intended to limit the invention and represent preferred dose ranges. However, the most preferred dosage will be tailored to the individual subject, as is understood and determinable by one of skill in the art, without undue experimentation. See, e.g., Berkow et al, eds., Merck Manual, 16th edition, Merck and Co., Rahway, NJ.
  • Effective amounts of a diagnostic/pharmaceutical compound or composition of the present invention are from about 0.001 ⁇ g to about 100 mg/kg body weight, administered at intervals of 4-72 hours, for a period of 2 hours to 5 years, or any range or value therein, such as 0.01-1.0, 1.0- 10, 10-50 and 50-100 mg/kg, at intervals of 1-4, 6-12, 12-24 and 24-72 hours, for a period of 0.5, 1.0-2.0, 2.0-4.0 and 4.0-7.0 days, or 1, 1-2, 2-4, 4-52 or more weeks, or 1, 2, 3-10, 10-20, 20-60 or more years, or any range or value therein.
  • Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions, which may contain auxiliary agents or excipients which are known in the art.
  • Pharmaceutical compositions such as tablets and capsules can also be prepared according to routine methods. See, e.g. , Berker, supra, Goodman, supra, Avery, supra and Ebadi, supra, which are entirely inco ⁇ orated herein by reference, including all references cited therein.
  • compositions comprising at least one type of sFv conjugate of the invention, or, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 types of sFv conjugates, ofthe present invention may be contained in an amount effective to achieve its intended pu ⁇ ose.
  • a pharmaceutical composition may contain suitable pharmaceutically acceptable carriers, such as excipients, carriers and/or auxiliaries which facilitate processing ofthe active compounds into preparations which can be used pharmaceutically.
  • compositions may also include suitable solutions for administration intravenously, subcutaneously, dermally, orally, mucosally or rectally, and contain from about 0.01 to 99 percent, preferably from about 20 to 75 percent of active component (i.e., the sFv) together with the excipient.
  • Pharmaceutical compositions for oral administration include tablets and capsules.
  • Compositions which can be administered rectally include suppositories. See, e.g., Berker, supra, Goodman, supra, Avery, supra and Ebadi, supra. Additional lipid and lipoprotein drug delivery systems that may be included herein are described more fully in Annals N Y. Acad. Sci. 507:775-88, 98-103, and 252-271, which disclosure is hereby inco ⁇ orated by reference.
  • compositions may also be formulated into orally administrable compositions containing one or more physiologically compatible carriers or excipients, and may be solid or liquid in form.
  • These compositions may, if desired, contain conventional ingredients such as binding agents, for example, syrups, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, such as lactose, mannitol, starch, calcium phosphate, sorbitol, cyclodextran, or methylcellulose; lubricants such as magnesium stearate, high molecular weight polymers such as polyethylene glycols, high molecular weight fatty acids such as stearic acid or silica; disintegrants such as starch; acceptable wetting agents as, for example, sodium lauryl sulfate.
  • binding agents for example, syrups, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone
  • the oral compositions may assume any convenient form, such as tablets, capsules, lozenges, aqueous or oily suspensions, emulsions, or dry products suitable for reconstitution with water or other liquid medium prior to use.
  • the liquid oral forms may, of course, contain flavors, sweeteners, preservatives such as methyl or propyl p-hydroxybenzoates; suspending agents such as sorbitol, glucose or other sugar syrup, methyl, hydroxymethyl, or carboxymethyl celluloses or gelatin; emulsifying agents such as lecithin or sorbitan monooleate or thickening agents.
  • Non-aqueous compositions may also be formulated which comprise edible oils as, for example, fish-liver or vegetable oils. These liquid compositions may conveniently be encapsulated in, for example, gelatin capsules in a unit dosage amount.
  • compositions according to the present invention may also be administered, if appropriate, either topically as an aerosol or, formulated with conventional bases as a cream or ointment.
  • compositions of the present invention can also be administered by inco ⁇ orating the active ingredient into colloidal carriers, such as liposomes.
  • colloidal carriers such as liposomes.
  • Liposome technology is well known in the art, having been described by Allison et al, Nature 252:252-254 (1974), and Dancy et al, J. Immunol. 720:1109-1113 (1978).
  • the frozen cell paste from three 10-liter fermentations (600-900 g) was thawed overnight at 4°C and gently resuspended at 4°C in 50 mM Tris-HCl, 1.0 mM EDTA, 100mM KCl, 0J mMPMSF,pH 8.0 (lysis buffer), using 10 liters of lysis buffer for every kilogram of wet cell paste.
  • the chilled mixture was passed three times through a Manton-Gaulin cell homogenizer to totally lyse the cells.
  • the cell homogenizer raised the temperature of the cell lysate to 25 ⁇ 5°C
  • the cell lysate was cooled to 5 + 2°C with a Lauda/Brinkman chilling coil after each pass. Complete lysis was verified by visual inspection under a microscope. The cell lysate was centrifuged at 24,300g for 30 minutes at 6°C using a
  • Sorvall RC-5B centrifuge The pellet containing the insoluble single-chain antigen-binding protein was retained, and the supernatant was discarded. The pellet was washed by gently scraping it from the centrifuge bottles and resuspending it in 5 liters of lysis buffer/kg of wet cell paste. The resulting 3.0- to 4.5-liter suspension was again centrifuged at 24,300g for 30 minutes at 6°C, and the supernatant was discarded. This washing ofthe pellet removes soluble E. coli proteins and can be repeated as many as five times. At any time during this washing procedure the material can be stored as a frozen pellet at -20°C. A substantial time saving in the washing steps can be accomplished by utilizing a Pellicon tangential flow apparatus equipped with 0.22- ⁇ m microporous filters, in place of centrifugation.
  • the washed pellet was solubilized at 4°C in freshly prepared 6 M guanidine hydrochloride, 50 mM Tris-HCl, 10 mM CaCl 2 , 50 mM KC1, pH 8.0 (dissociating buffer), using 9 ml/g of pellet. If necessary, a few quick pulses from a Heat Systems Ultrasonics tissue homogenizer can be used to complete the solubilization. The resulting suspension was centrifuged at 24,300g for 45 minutes at 6°C and the pellet was discarded. The optical density of the supernatant was determined at 280 nm and if the OD 2g0 was above 30, additional dissociating buffer was added to obtain an OD 280 of approximately 25.
  • the supernatant was slowly diluted into cold (4-7°C) refolding buffer (50 mM Tris-HCl, 10 mM CaCl 2 , 50 mM KC1, pH 8.0) until a 1 :10 dilution was reached (final volume 10 - 20 liters). Re-folding occurs over approximately eighteen hours under these conditions. The best results are obtained when the GuHCl extract is slowly added to the refolding buffer over a two hour period, with gentle mixing.
  • the solution was filtered through a 0.2 ⁇ m Millipore Millipak 200. This filtration step may be optionally preceded by a centrifugation step.
  • the filtrate was concentrated to 1 to 2 liters using an .Amicon spiral cartridge with 10,000 MWCO cartridge, again at 4°C.
  • the concentrated crude antigen-binding protein sample was dialyzed against Buffer G (60 mM MOPS, 0.5 mM Ca acetate, pH 6.0 - 6.4) until the conductivity was lowered to that of Buffer G.
  • Buffer G 60 mM MOPS, 0.5 mM Ca acetate, pH 6.0 - 6.4
  • the sample was then loaded on a 21.5 x 250-mm polyaspartic acid PolyCAT A column, manufactured by Poly LC of Columbia, Maryland. If more than 60 mg of protein is loaded on this column, the resolution begins to deteriorate; thus, the concentrated crude sample often must be divided into several PolyCAT A runs.
  • Most antigen-binding proteins have an extinction coefficient of about 2.0 ml mg 'cm "1 at 280 nm and this can be used to determine protein concentration.
  • the antigen-binding protein sample was eluted from the PolyCAT A column with a 50-min linear gradient from Buffer G to Buffer H (60 mM MOPS, 20 mM Ca Acetate, pH 7.5 - 8.0). Most of the single-chain proteins elute between 20 and 26 minutes when this gradient is used. This corresponds to an eluting solvent composition of approximately 70% Buffer G and 30% Buffer H. Most ofthe bivalent antigen-binding proteins elute later than 45 minutes, which correspond to over 90%) Buffer H.
  • a sample of CC 49/212 single chain antigen binding molecule (MW 27000) dissolved in KPO 4 /NaCl buffer (pH 7.2) was obtained as described in Example 2.
  • the protein was found to be pure using SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) and size exclusion chromatography. The concentration of the protein was 0.79 mg/ml. It was further concentrated to at least 2 mg/ml using an Amicon concentrator with a 10,000 dalton nominal size cut-off, i.e., anything greater than 10K is retained.
  • the modification reaction i.e., the coupling of the SC-PEG to the CC 49/212, was carried out in 50 mM KPO 4 , 150 mM NaCl buffer, which was the storage buffer the protein was supplied in. The pH was raised from 7.2 to 7.5. SC-PEG (MW 5,000) was added in a 50x molar excess to protein. At specific time intervals, the coupling reaction was terminated by the addition of a 5 Ox molar excess of glycine and the extent and progress of the coupling reaction was checked as a function of time using both size exclusion chromatography using a DuPont Zorbax 250 column and SDS-PAGE.
  • the samples were checked for degree of modification by size exclusion chromatography. After concentrating the samples, the residual amine concentration on the protein was determined by titration with trinitrobenzene sulfonate and the percentage of amine groups that had reacted with the SC-PEG (the "% modification") was calculated from the results.
  • the CC49 monoclonal antibody was developed by Dr. Jeffrey Schlom's group, Laboratory of Tumor Immunology and Biology, National Cancer Institute. It binds specifically to the pan-carcinoma tumor antigen TAG-72. See Muraro, R. et al, Cancer Research 48: 4588-4596 (1988).
  • Figure 1 is a graphical representation of three competition ELISA's in which unlabeled PEG modified CC49/212 single-chain Fv (closed squares), CC49/212 single-chain Fv (open squares), CC49 IgG (open circles), and MOPC- 21 IgG (+) competed against a CC49 IgG radiolabeled with 125 I for binding to the TAG-72 antigen on a human breast melanoma extract.
  • MOPC-21 is a control antibody that does not bind to TAG-72 antigen.
  • Methoxypolyethylene glycol (m-PEG) was obtained from Union Carbide. The solvents were obtained from Aldrich Chemical of Milwaukee, Wisconsin. The methoxypoly(ethylene glycol)-N-succinimidyl carbonate (SC-PEG) was prepared as described in U.S. Patent No. 5,122,614, using m-PEG having a molecular weight of about 5,000. Each of the products prepared in Examples 5-10 was confirmed structurally by carbon- 13 NMR.
  • the branched polymer, U-PEG-OH was prepared by adding 100 mg ( 1 J mmol) of 1 ,3-diamino-2-propanol to a solution of 10.0 g (2 mmol) of SC-PEG in 50 mL of methylene chloride. The mixture was stirced for 18 hours at room temperature then filtered. Excess solvent was removed by distillation in vacuo. The residue was recrystallized from 2-propanol to yield 7J g of product (70% yield).
  • the compound of Example 5 was activated with p-nitrophenyl chloroformate.
  • 5.0g (0.5 mmol) of U-PEG-OH was azeotropically dried by refiuxing in 75 mL of toluene for 2 hours, resulting in the removal of 25 mL of solvent/water.
  • the reaction mixture was cooled to 30 ° C, followed by the addition of 120 mg (0.6 mmol) of p-nitrophenyl chloroformate and 50 mg (0.6 mmol) of pyridine.
  • the resulting mixture was stined for two hours at 45 °C, followed by stirring overnight at room temperature.
  • reaction mixture was then filtered through CELITETM , followed by removal ofthe solvent from the filtrate by distillation in vacuo. The residue was recrystallized from 2-propanol to yield 4.2 g (81% yield) ofthe product.
  • the U-PNP-PEG of Example 6 was reacted with N-hydroxysuccinimide to form the succinimidyl carbonate ester of U-PEG.
  • a solution containing 5.0 g (0.5 mmol) of the U-PNP-PEG, 0.6 g (5 mmol) of N-hydroxysuccinimide and 0J3 g (1 mmol) of diisopropylethylamine in 40 ml of methylene chloride was refluxed for 18 hours. The solvent was then removed by distillation in vacuo, and the residue was recrystallized from 2-propanol to yield 4.2 g ofthe succinimidyl carbonate ester (82% yield).
  • Example 8 Preparation of XU-PEG-OH
  • This branched polymer was prepared by reacting the U-PNP-PEG of Example 6 with 2-(2-aminoethoxy) ethanol (i.e., the amino alcohol was reacted with the p-nitrophenyl carbonate).
  • the recrystallized product yield was 86%.
  • Example 8 The compound of Example 8 was functionalized with p-nitrophenyl carbonate as in Example 6. The recrystallized product yield was 83%.
  • succinimidyl carbonate derivative of compound prepared in Example 8 was prepared according to the process described in Example 7, by reacting N-hydroxysuccinimide with the p-nitrophenyl carbonate derivative of Example 9.
  • the recovered product yield was 84%.
  • a sample containing CC49/218 was desalted on a PD-10 column in a buffer consisting of OJM sodium phosphate, pH 8.0. .An equimolar amount of
  • the assay was performed as in Example 4 above using SC-PEG modified CC49/218 and XUS-PEG modified CC49/218 along with the appropriate controls. The results are shown in Figure 4 and in Table 4 below.
  • the affinity ofthe SC-PEG modified CC49-SCA was within about 8 to 10 fold of the native CC49-SCA and the affinity the XUS-PEG modified CC49-SCA was within about 4 to 5 fold ofthe native CC49-SCA.
  • Samples #049304 and #049303 were PEG modified, whiles samples #04901 and #049302 were unmodified CC49/218 isolated from the reaction mixtures.
  • CC49-SCA dimers and trimers of CC49-SCA have been made.
  • CC49-SCA was modified with bifunctional PEG as follows: 2 ml of CC49- SCA in phosphate buffered saline (25 mM sodium phosphate, pH 7.3, 0J5 M NaCl) at a concentration of 1.5 mg/ml was modified as follows.
  • Bifunctional PEG polyethylene glycol with reactive SC at both terminal ends
  • 1.887 mg (powder) was dissolved in OJ ml of MOPS (3-[N-mo ⁇ holino]propane)-sulfonic acid) buffered at pH 7.3. This PEG solution was added to CC49-SCA solution within 10 seconds of dissolution.
  • the multimers were assayed for binding affinity using the following assay which was modified method described in B Friquet et al. J. of Immunology Methods, 77:305-319 ( 1985). Briefly, various amounts of a given modified single chain antibody were mixed with various amounts ofthe antigen mucin in PBS (phosphate buffered saline). The binding reaction was allowed to reach equilibration for at least 24 hours at 4°C. At the end of the incubation, the unbound CC49-SCA fractions were assayed by ELISA, while the bound fractions were washed away.
  • the total amount of free CC49-SC A was determined by the ELISA using the CC49-SCA sample pre-incubated in the absence ofthe antigen mucin.
  • the bound antibody was determined by subtraction ofthe free (unbound) amount from the total amount as determined by ELISA. Since the total amount ofthe CC49-SCA was known, it was also used as its own standard curve. Note that each type of CC49-SCA had its own reference control. In essence, the protocol was measuring the unbound amount of a particular version of PEG- CC49-SC A as a result ofthe binding to the antigen. Although PEG may affect the detection reagent in ELISA, this was well contained in the standard references. Therefore, the amount measured was not due to the difference of various PEG on the various versions of PEG-CC49-SCA.
  • PEG increases. As more PEG is attached to the protein, the circulation time increases. However, attachment of a few strands of high molecular weight PEG gives a better increase in circulation half-life than multiple stands of lower molecular weight PEG.
  • the linker is an SC-bond, Flan-bond, hydrazine bond, or TPC, there was no significant change in the circulation half-life. Therefore, the chemical bonds of the linkers, if not releasable, do not affect the circulation half-life. In addition, the PEG remains attached to the protein during the observable time.
  • the PK modeling was following a one-compartment, i.v.-bolus model.
  • the circulatory half-life is the time for the drug concentration in the serum to reduce to one half after equilibration is reached (about 2 min.)
  • AUC area under the curve, is the integral ofthe drug blood level over time from zero to last measurement, and is a measure of quantity of drug absorbed and in the body.
  • Mean residence time is the average amount of time a drug remains in the compartment.
  • the Flan-bond PEG was prepared as taught in U.S. Patent 5,405,877.
  • a competition binding assay of biotinylated CC49-SCA with various PEGylated CC49-SCA proteins was performed using an ELISA ofthe biotinylated CC49-SCA as detected by horseradish-peroxidase conjugated with streptoavidin
  • biotinylated CC49-SCA and PEG-CC49-SCA sample were mixed at various ratios for competition of binding to the antigen mucin on a surface.
  • the amount of biotinylated-CC49-SCA bound to antigen was then measured by SAN-HRP, which would not detect the PEG-modified CC49- SCA.
  • PEG-CC49-SCA is a reflection ofthe relative affinity ofthe two forms of CC49- SCA for the antigen.
  • the affinity ranking obtained was as follows:
  • SC2 is PEG SC2000-CC49-SCA
  • GC is glyco-CC49-SCA
  • Bio.CC49-SCA is the biotinylated CC49-SCA
  • CC12 is the PEG-SC12,000-CC49-SCA
  • F5 is PEG-Flan-5000-CC49-SCA
  • HZ is CC49-SCA highly PEGylated on the carboxyl groups with MW 5000 hydrazine-PEG
  • PG is PEG-glyco-CC49-SCA and is highly PEGylated on the carbohydrate with MW 5000 hydrazine PEG
  • C20 is PEG-SC20000-CC49-SC A.
  • ADDRESSEE STERNE, KESSLER, GOLDSTEIN & FOX P.L.L.C.
  • TELECOMMUNICATION INFORMATION (A) TELEPHONE: 202-371-2600 (B) TELEFAX: 202-371-2540
  • GGT AAT CAA AAG AAC TAC TTG GCC TGG TAC CAG CAG AAA CCA GGG CAG 144
  • TCT CCT AAA CTG CTG ATT TAC TGG GCA TCC GCT AGG GAA TCT GGG GTC 192
  • ATC AGC TGT GTG AAG ACT GAA GAC CTG GCA GTT TAT TAC TGT CAG CAG 288 lie Ser Cys Val Lys Thr Glu Asp Leu Ala Val Tyr Tyr Cys Gin Gin
  • Glu Trp lie Gly Tyr Phe Ser Pro Gly Asn Asp Asp Phe Lys Tyr Asn 180 185 190
  • GGT AAT CAA AAG AAC TAC TTG GCC TGG TAC CAG CAG AAA CCA GGG CAG 144
  • TCT CCT AAA CTG CTG ATT TAC TGG GCA TCC GCT AGG GAA TCT GGG GTC 192

Abstract

The present invention relates to the chemical modification of single chain polypeptides by means of covalent attachment of strands of poly(ethylene glycol) PEG and similar poly(alkylene oxides) to single chain polypeptide binding molecules that have the three dimensional folding and, thus, the binding ability and specificity, of the variable region of an antibody. Such preparations of modified single chain polypeptide binding molecules have reduced immugenicity and antigenicity as well as having a longer halflife in the bloodstream as compared to the parent polypeptide. These beneficial properties of the modified single chain polypeptide binding molecules make them very useful in a variety of therapeutic applications. The invention also relates to multivalent antigen-binding molecules capable of PEGylation. Compositions of, genetic constructions for, methods of use, and methods for producing PEGylated antigen-binding proteins are disclosed.

Description

Polyalkylene Oxide-Modified Single Chain Polypeptides
Background of the Invention
1. Field of the Invention
The present invention relates to the chemical modification of single chain polypeptides by means of covalent attachment of strands of poly(ethylene glycol) PEG and similar poly(alkylene oxides) to single chain polypeptide binding molecules that have the three dimensional folding and, thus, the binding ability and specificity, of the variable region of an antibody. Such preparations of modified single chain polypeptide binding molecules have reduced immugenicity and antigenicity as well as having a longer halflife in the bloodstream as compared to the parent polypeptide. These beneficial properties of the modified single chain polypeptide binding molecules make them very useful in a variety of therapeutic applications. The invention also relates to multivalent antigen-binding molecules capable of PEGylation. Compositions of, genetic constructions for, methods of use, and methods for producing PEGylated antigen-binding proteins are disclosed.
2. Description of Related Art
.Antibodies are proteins generated by the immune system to provide a specific molecule capable of complexing with an invading molecule, termed an antigen. Natural antibodies have two identical antigen-binding sites, both of which are specific to a particular antigen. The antibody molecule "recognizes" the antigen by complexing its antigen-binding sites with areas of the antigen termed epitopes. The epitopes fit into the conformational architecture of the antigen- binding sites of the antibody, enabling the antibody to bind to the antigen.
The antibody molecule is composed of two identical heavy and two identical light polypeptide chains, held together by interchain disulfide bonds. The remainder of this discussion on antibodies will refer only to one pair of light/heavy chains, as each light/heavy pair is identical. Each individual light and heavy chain folds into regions of approximately 110 amino acids, assuming a conserved three- dimensional conformation. The light chain comprises one variable region (VL) and one constant region (CL), while the heavy chain comprises one variable region (NH) and three constant regions (CH1, CH2 and CH3). Pairs of regions associate to form discrete structures. In particular, the light and heavy chain variable regions associate to form an "Fv" area which contains the antigen-binding site. The constant regions are not necessary for antigen binding and in some cases can be separated from the antibody molecule by proteolysis, yielding biologically active (i.e., binding) variable regions composed of half of a light chain and one quarter of a heavy chain.
Further, all antibodies of a certain class and their Fab fragments (i.e., fragments composed of NL, CL, VH, and CH1) whose structures have been determined by x-ray crystallography show similar variable region structures despite large differences in the sequence of hypervariable segments even when from different animal species. The immunoglobulin variable region seems to be tolerant towards mutations in the antigen-binding loops. Therefore, other than in the hypervariable regions, most of the so-called "variable" regions of antibodies, which are defined by both heavy and light chains, are, in fact, quite constant in their three dimensional arrangement. See for example, Huber, R., Science 233:702-703 (1986)).
Recent advances in immunobiology, recombinant DΝA technology, and computer science have allowed the creation of single polypeptide chain molecules that bind antigen. These single-chain antigen-binding molecules ("SCA") or single-chain variable fragments of antibodies ("sFv") incorporate a linker polypeptide to bridge the individual variable regions, VL and NH, into a single polypeptide chain. A description of the theory and production of single-chain antigen-binding proteins is found in Ladner et al, U.S. Patent Νos. 4,946,778, 5,260,203, 5,455,030 and 5,518,889. The single-chain antigen-binding proteins produced under the process recited in the above U.S. patents have binding specificity and affinity substantially similar to that of the corresponding Fab fragment. A computer-assisted method for linker design is described more particularly in Ladner et al. , U.S. Patent Νos. 4,704,692 and 4,881 J 75, and WO 94/12520. The in vivo properties of sFv (SCA) polypeptides are different from MAbs and antibody fragments. Due to their small size, sFv (SCA) polypeptides clear more rapidly from the blood and penetrate more rapidly into tissues (Milenic, D.E. et al. , Cancer Research 51 :6363 -6371 ( 1991 ); Colcher et al. , J. Natl. Cancer Inst. 82:1191 (1990); Yokota et al., Cancer Research 52:3402 (1992)). Due to lack of constant regions, sFv (SCA) polypeptides are not retained in tissues such as the liver and kidneys. Due to the rapid clearance and lack of constant regions, sFv (SCA) polypeptides will have low immunogenicity. Thus, sFv (SCA) polypeptides have applications in cancer diagnosis and therapy, where rapid tissue penetration and clearance, and ease of microbial production are advantageous. A multivalent antigen-binding protein has more than one antigen-binding site. A multivalent antigen-binding protein comprises two or more single-chain protein molecules. Enhanced binding activity, di- and multi-specific binding, and other novel uses of multivalent antigen-binding proteins have been demonstrated. See, Whitlow, M., et al., Protein Engng. 7:1017-1026 (1994); Hoogenboom, H.R., Nature Biotech. 75:125-126 (1997); and WO 93/11161.
Ladner et al. also discloses the use of the single chain antigen binding molecules in diagnostics, therapeutics, in vivo and in vitro imaging, purifications, and biosensors. The use of the single chain antigen binding molecules in immobilized form, or in detectably labeled forms is also disclosed, as well as conjugates of the single chain antigen binding molecules with therapeutic agents, such as drugs or specific toxins, for delivery to a specific site in an animal, such as a human patient.
Whitlow et al. {Methods: A Companion to Methods in Enzymology 2(2) : 97- 105 (June, 1991 )) provide a good review of the art of single chain antigen binding molecules and describe a process for making them.
In U.S. Patent 5,091,513, Huston et al. discloses a family of synthetic proteins having affinity for preselected antigens. The contents of U.S.Patent 5,091,513 are incorporated by reference herein. The proteins are characterized by one or more sequences of amino acids constituting a region that behaves as a biosynthetic antibody binding site (BABS). The sites comprise (1 ) noncovalently associated or disulfide bonded synthetic VH and VL regions, (2) NH-VL or VL-NH single chains wherein the VH and VL are attached to a polypeptide linker, or (3) individual VH or VL domains. The binding domains comprises complementarity determining regions (CDRs) linked to framework regions (FRs), which may be derived from separate immunoglobulins.
U.S. Patent 5,091,513 also discloses that three subregions (the CDRs) of the variable domain of each of the heavy and light chains of native immunoglobulin molecules collectively are responsible for antigen recognition and binding. These CDRs consist of one of the hypervariable regions or loops and of selected amino acids or amino acid sequences disposed in the framework regions that flank that particular hypervariable region. It is said that framework regions from diverse species are effective in maintaining CDRs from diverse other species in proper conformation so as to achieve true immunochemical binding properties in a biosynthetic protein. U.S. Patent 5 ,091 ,513 includes a description of a chimeric polypeptide that is a single chain composite polypeptide comprising a complete antibody binding site. This single chain composite polypeptide is described as having a structure patterned after tandem VH and VL domains, with a carboxyl terminal of one attached through an amino acid sequence to the amino terminal of the other. It thus comprises an amino acid sequence that is homologous to a portion of the variable region of an immunoglobulin heavy chain (NH) peptide bonded to a second amino acid sequence that was homologous to a portion of the variable region of an immunoglobulin light chain (VL).
The covalent attachment of strands of a polyalkylene glycol to a polypeptide molecule is disclosed in U.S. Patent No.4, 179,337 to Davis et al. , as well as in Abuchowski and Davis "Enzymes as Drugs," Holcenberg and Roberts, Eds., pp. 367-383, John Wiley .and Sons, New York (1981). These references disclosed that proteins and enzymes modified with polyethylene glycols have reduced immunogenicity and antigenicity and have longer lifetimes in the bloodstream, compared to the parent compounds. The resultant beneficial properties of the chemically modified conjugates are very useful in a variety of therapeutic applications.
Although amino acid sequences such as the single chain polypeptides described above, and fusion proteins thereof, have not been associated with significant antigenicity in mammals, it has been desirable to prolong the circulating life and even further reduce the possibility of an antigenic response. The relatively small size of the polypeptides and their delicate structure/activity relationship, however, have made polyethylene glycol modification difficult and unpredictable. Most importantly, it was unknown how to modulate retained activity of the polypeptides after conjugation with polymers, such as PEG.
To effect covalent attachment of polyethylene glycol (PEG) or polyalkalene oxides to a protein, the hydroxyl end groups of the polymer must first be converted into reactive functional groups. This process is frequently referred to as "activation" and the product is called "activated PEG" or activated polyalkylene oxide. Methoxy poly(ethylene glycol) (mPEG), capped on one end with a functional group, reactive towards amines on a protein molecule, is used in most cases.
The activated polymers are reacted with a therapeutic agent having nucleophilic functional groups that serve as attachment sites. One nucleophilic functional group commonly used as an attachment site is the e-amino groups of lysines. Free carboxylic acid groups, suitably activated carbonyl groups, oxidized carbohydrate moieties and mercapto groups have also been used as attachment sites.
The hydroxyl group of PEG has been activated with cyanuric chloride and the resulting compound is then coupled with proteins (Abuchowski et al, J. Biol. Chem. 252:3578 (1977); Abuchowski & Davis, supra (1981)). However, there are disadvantages in using this method, such as the toxicity of cyanuric chloride and its non-specific reactivity for proteins having functional groups other than amines, such as free essential cysteine or tyrosine residues. In order to overcome these and other disadvantages, alternative activated
PEGs, such as succinimidyl succinate derivatives of PEG ("SS-PEG"), have been introduced (Abuchowski et al, Cancer Biochem. Biophys. 7:175-186 (1984)). SS-PEG reacts quickly with proteins (30 minutes) under mild conditions yielding active yet extensively modified conjugates. Zalipsky, in U.S. Patent No.5, 122,614, discloses poly(ethylene glycol)-N- succinimide carbonate and its preparation. This form of the polymer is said to react readily with the amino groups of proteins, as well as low molecular weight peptides and other materials that contain free amino groups.
Other linkages between the amino groups of the protein, and the PEG are also known in the art, such as urethane linkages (Veronese et al, Appl. Biochem. Biotechnol 77:141-152 (1985)), carbamate linkages (Beauchamp et al, Analyt. Biochem. 131:25-33 (1983)), and others.
Suzuki et al. {Biochimica et Biophysica Acta, 788: 248-255 (1984)) covalently couples immunoglobulin G (IgG) to poly(ethylene glycol) that has previously been activated by cyanuric chloride. The coupled IgG was studied for physicochemical and biological properties such as molecular structure, size- exclusion chromatographic behavior, surface activity, interfacial aggregability, heat aggregability inducing nonspecific complement activation, and antigen- binding activity. The poly(ethylene glycol) coupling to IgG increased the apparent Stokes' radius and the surface activity of IgG and stabilized IgG on heating and/or on exposure to interfaces, while no structural denaturation of IgG was observed. The suppressed nonspecific aggregability was interpreted mainly by difficulty in association between the modified IgG molecules. These results indicated the use of the poly(ethylene glycol)-coupled IgG as an intravenous preparation and also as an additive stabilizing intact IgG for intravenous use.
Sharp et al. {Analytical Biochemistry 154: 110-117 (1986)) investigated the possibility of producing biospecific affinity ligands for separating cells in two polymer aqueous phase systems on the basis of cell surface antigens. Rabbit anti- human erythrocyte IgG was reacted with cyanuric chloride-activated monomethyl poly(ethylene glycol) fractions (molecular weights approximately 200, 1900, and 5000) at various molar ratios of PEG to protein lysine groups. The partition coefficient of the protein in a Dextran PEG two phase system increased with increasing degree of modification and increasing PEG molecular weight. There was a concomitant loss in ability to agglutinate human erythrocytes. Tullis, in U.S. Patent No.4,904,582, describes oligonucleotide conjugates wherein the oligonucleotides are joined through a linking arm to a hydrophobic moiety, which could be a polyalkyleneoxy group. The resulting conjugates are said to be more efficient in membrane transport, so as to be capable of crossing the membrane and effectively modulating a transcriptional system. In this way, the compositions can be used in vitro and in vivo, for studying cellular processes, protecting mammalian hosts from pathogens, and the like.
Excessive polymer conjugation and/or conjugation involving a therapeutic moietie's active site where groups associated with bioactivity are found, however, often result in loss of activity and, thus, therapeutic usefulness. This is often the case with lower molecular weight peptides which have few attachment sites not associated with bioactivity. For example, Benhar et al. {Bioconjugate Chem. 5:321-326 (1994)) observed that PEGylation of a recombinant single-chain immunotoxin resulted in the loss of specific target immunoreactivity of the immunotoxin. The loss of activity of the immunotoxin was the result of PEG conjugation at two lysine residues within the antibody-combining region of the immunotoxin. To overcome this problem, Benhar et al. replaced these two lysine residues with arginine residues and were able to obtain an active immunotoxin that was 3-fold more resistant to inactivation by derivatization. aAnother suggestion for overcoming these problems discussed above is to use longer, higher molecular weight polymers. These materials, however, are difficult to prepare and expensive to use. Further, they provide little improvement over more readily available polymers.
Another alternative suggested is to attach two strands of polymer via a triazine ring to amino groups of a protein. See, for example, Enzyme 26:49-53 (1981) and Proc. Soc. Exper. Biol. Med, 188:364-369 (1988). However, triazine is a toxic substance that is difficult to reduce to acceptable levels after conjugation. Thus, non-triazine-based activated polymers would offer substantial benefits to the art.
Summary of the Invention
The present invention relates to polyalkylene oxide/amino acid sequence conjugates and processes for preparing them. Suitable amino acid sequences are peptides, such as, single chain polypeptides having binding affinity for an antigen, for example, those described by Ladner et al. in U.S. Patent No. 4,946,778 and Huston et al. in U.S. Patent No. 5,091,513. More particularly, the present invention relates to a physiologically active, substantially non-immunogenic polypeptide conjugate containing at least one polyalkylene oxide strand coupled to a single chain polypeptide having binding affinity for an antigen. The single chain polypeptide includes:
(a) a first polypeptide comprising the binding portion of the light chain variable region of an antibody;
(b) a second polypeptide comprising the binding portion of the heavy chain variable region of an antibody; and (c) at least one peptide linker linking said first and second polypeptides (a) and (b) into said single chain polypeptide having binding affinity for the antigen. In another aspect, the present invention relates to a process for preparing physiologically active, substantially non-immunogenic polypeptide compositions. The process includes coupling a polyalkylene oxide to a single chain polypeptide having the attributes described above. Preferably, the poly(alkylene oxides) used herein are poly(ethylene glycols) that have been activated for coupling to the target polypeptide. The invention is also directed to a single-chain antigen-binding polypeptide
-polyalkylene oxide conjugate, comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within a range of about one-fold to about ten-fold of the antigen binding affinity of the native, unconj ugated foim of the single-chain antigen-binding polypeptide.
The invention is also directed to a single-chain antigen-binding polypeptide -polyalkylene oxide conjugate, comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about ten-fold of the antigen binding affinity of the native, unconjugated form of the single-chain antigen-binding polypeptide.
The invention is also directed to a single-chain antigen-binding polypeptide -polyalkylene oxide conjugate, comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and (c) a peptide linker linking the first and second polypeptides (a) and
(b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about five-fold of the antigen binding affinity of the native, unconjugated form of the single-chain antigen-binding polypeptide. The invention is also directed to a single-chain antigen-binding polypeptide
-polyalkylene oxide conjugate, comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about two-fold of the antigen binding affinity of the native, unconjugated form of the single-chain antigen-binding polypeptide.
The invention is also directed to a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; (b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue wherein the Cys residue is capable of polyalkylene oxide conjugation and the Cys residue is located at a position selected from the group consisting of (i) the amino acid position 11 , 12, 13 , 14 or 15 of the light chain variable region; (ii) the amino acid position 77, 78 or 79 of the light chain variable region; (iii) the amino acid position 11, 12, 13, 14 or 15 of the heavy chain variable region; (iv) the amino acid position 82B, 82C or 83 of the heavy chain variable region; (v) any amino acid position of the peptide linker; (vi) adjacent to the C-terminus of polypeptide
(a) or (b); and (vii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
The invention is also directed to a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; (b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and
(b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least three consecutive Lys residues wherein the consecutive Lys residues are capable of polyalkylene oxide conjugation and any one of the consecutive Lys residues is located at a position selected from the group consisting of (i) any amino acid position of the peptide linker; (ii) adjacent to the C-terminus of polypeptide (a) or (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen. These consecutive lysine residues in the sFv (SCA) protein (i.e., oligo-lysine sFv) generate a "hot spot" for polyalkylene oxide conjugation.
The invention is also directed to a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising: (a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least two consecutive Cys residue wherein the consecutive Cys residues are capable of polyalkylene oxide conjugation and any one of the consecutive Cys residues is located at a position selected from the group consisting of (i) any amino acid position of the peptide linker; (ii) adjacent to the C-terminus of polypeptide (a) or (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen. These consecutive cysteine residues in the sFv (SCA) protein (i.e., oligo-cysteine sFv) generate a "hot spot" for polyalkylene oxide conjugation. The invention is further directed to a genetic sequence encoding a single- chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; (b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue wherein the Cys residue is capable of polyalkylene oxide conjugation and the Cys residue is located at a position selected from the group consisting of (i) the amino acid position 11 , 12, 13, 14 or 15 ofthe light chain variable region; (ii) the amino acid position 77, 78 or 79 ofthe light chain variable region; (iii) the amino acid position 11, 12, 13, 14 or 15 ofthe heavy chain variable region; (iv) the amino acid position 82B, 82C or 83 ofthe heavy chain variable region; (v) any amino acid position ofthe peptide linker; (vi) adjacent to the C-terminus of polypeptide
(a) or (b); and (vii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen. The invention is further directed to a genetic sequence encoding a single- chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; (b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and
(b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least three consecutive Lys residue wherein the consecutive Lys residues are capable of polyalkylene oxide conjugation and any one of the consecutive Lys residues is located at a position selected from the group consisting of (i) any amino acid position ofthe peptide linker; (ii) adjacent to the C-terminus of polypeptide (a) or (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen. These consecutive lysine residues in the sFv (SCA) protein (i.e., oligo-lysine sFv) generate a "hot spot" for polyalkylene oxide conjugation.
The invention is further directed to a genetic sequence encoding a single- chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising: (a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and (c) a peptide linker linking the first and second polypeptides (a) and
(b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least two consecutive Cys residue wherein the consecutive Cys residues are capable of polyalkylene oxide conjugation and any one of the consecutive Cys residues is located at a position selected from the group consisting of (i) any amino acid position ofthe peptide linker; (ii) adjacent to the C-terminus of polypeptide (a) or (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen. These consecutive cysteine residues in the sFv (SCA) protein (i.e., oligo-cysteine sFv) generate a "hot spot" for polyalkylene oxide conjugation.
The genetic sequence may be DNA or RNA.
The invention is directed to a replicable cloning or expression vehicle comprising the above described DNA sequence. The invention is also directed to such vehicle which is a plasmid. The invention is further directed to a host cell transformed with the above described DNA. The host cell may be a bacterial cell, a yeast cell or other fungal cell, an insect cell or a mammalian cell line. A preferred host is Pichia pastor is.
The invention is directed to a method of producing a single-chain antigen- binding polypeptide capable of polyalkylene oxide conjugation, comprising: (a) providing a first genetic sequence encoding a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) providing a second genetic sequence encoding a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and (c) linking the first and second genetic sequences (a) and (b) with a third genetic sequence encoding a peptide linker into a fourth genetic sequence encoding a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue wherein the Cys residue is capable of polyalkylene oxide conjugation and the Cys residue is located at a position selected from the group consisting of (i) the amino acid position 11 , 12, 13 , 14 or 15 of the light chain variable region; (ii) the amino acid position 77, 78 or 79 ofthe light chain variable region; (iii) the amino acid position 11, 12, 13, 14 or 15 of the heavy chain variable region; (iv) the amino acid position 82B, 82C or 83 ofthe heavy chain variable region; (v) any amino acid position ofthe peptide linker; (vi) adjacent to the C-terminus of polypeptide
(a) or (b); and (vii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen; (d) transforming a host cell with the fourth genetic sequence encoding a single-chain antigen-binding polypeptide of (c); and
(e) expressing the single-chain antigen-binding polypeptide of (c) in the host, thereby producing a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation. The invention is further directed to a multivalent single-chain antigen- binding protein, comprising two or more single-chain antigen-binding polypeptides, each single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; (b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and
(b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue wherein the Cys residue is capable of polyalkylene oxide conjugation and the Cys residue is located at a position selected from the group consisting of (i) the amino acid position 11 , 12, 13, 14 or 15 ofthe light chain variable region; (ii) the amino acid position 77, 78 or 79 ofthe light chain variable region; (iii) the amino acid position 11, 12, 13, 14 or 15 ofthe heavy chain variable region; (iv) the amino acid position 82B, 82C or 83 ofthe heavy chain variable region; (v) any amino acid position ofthe peptide linker; (vi) adjacent to the C-terminus of polypeptide (a) or (b); and (vii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen. In the above described embodiments ofthe invention, the Cys polyalkylene oxide conjugation sequence may be capable of attaching a polyalkylene oxide moiety and the Cys residue is located at a position selected from the group consisting of (i') the amino acid position 77 ofthe light chain variable region; (ii') the amino acid position 82B ofthe heavy chain variable region; (iii') the amino acid position 3 ofthe peptide linker; (iv') adj acent to the C-terminus of polypeptide (a) or (b); (v') N-terminus and C-terminus; and (vi') combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
In the above described embodiments of the invention, the oligo-Lys polyalkylene oxide conjugation sequence may be capable of attaching a polyalkylene oxide moiety at the oligo-Lys residues located adjacent to the C- terminus ofthe protein, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
In the above described embodiments ofthe invention, the C-terminus of the second polypeptide (b) may be the native C-terminus. The C-terminus ofthe second polypeptide (b) may comprise a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe second polypeptide are sufficient for the polyalkylene oxide conjugated polypeptide to be capable of binding an antigen. The C-terminus of the second polypeptide may comprise an addition of one or plurality of amino acid residue(s), such that the polyalkylene oxide conjugated polypeptide is capable of binding an antigen.
In a preferred embodiment ofthe invention, the first polypeptide (a) may comprise the antigen binding portion ofthe variable region of an antibody light chain and the second polypeptide (b) comprises the antigen binding portion ofthe variable region of an antibody heavy chain.
The invention is also directed to a method of detecting an antigen suspected of being in a sample, comprising:
(a) contacting the sample with the polyalkylene oxide conjugated polypeptide or protein ofthe invention, wherein the polyalkylene oxide conjugated polypeptide is conjugated to one or plurality of detectable label molecule(s), or conjugated to a carrier having one or plurality of detectable label molecule(s) bound to the carrier; and
(b) detecting whether the polyalkylene oxide conj ugated single-chain antigen-binding polypeptide has bound to the antigen.
The invention is further directed to a method of imaging the internal structure of an animal, comprising administering to the animal an effective amount of the polyalkylene oxide conjugated polypeptide or protein of the invention, wherein the polyalkylene oxide conjugated polypeptide is conjugated to one or plurality of detectable label or chelator molecule(s), or conjugated to a carrier having one or plurality of detectable label or chelator molecule(s) bound to the carrier, and measuring detectable radiation associated with the animal. Animal includes human and nonhuman.
The invention is also directed to a method for treating a targeted disease, comprising administering an effective amount of a composition comprising the polyalkylene oxide conjugated polypeptide or protein of the invention and a pharmaceutically acceptable carrier vehicle, wherein the polyalkylene oxide conjugated polypeptide is conjugated to one or plurality of bioactive molecules, such as peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, boron addend or radioisotope molecule(s), or conjugated to a carrier having one or plurality of peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, boron addend or radioisotope molecule(s) bound to the carrier.
Brief Description ofthe Drawings
Figure 1 is a graphical representation of three competition ELISA's in which unlabeled PEG modified CC49/212 SCA (closed squares), CC49/212 SCA (open squares), CC49 IgG (open circles), and MOPC-21 IgG (+) competed against a CC49 IgG radiolabeled with 125I for binding to the TAG-72 antigen on a human breast carcinoma extract. Figure 2 shows the DNA and protein sequence of CC49/218 SCA which has four engineered cysteine residues at the positions indicated by the codons underlined and marked by an asterisk. Also highlighed are the CDR sequences (double underlined) and the 218 linker (underlined and labeled). In addition, there are four natural cysteine residues in the protein which are involved in two disulfide bonds. These are not underlined. The four engineered cysteine residues occur independently in four different mutants currently, but may be combined in the exact four-mutant codon version shown in this figure.
Figure 3 shows the DNA and protein sequence of CC49/218 SCA with an engineered oligo-lysine C-terminal tail segment. The eight new lysine residues were genetically engineered at a BstEII site and are shown underlined and marked with asterisks. Also highlighted are the CDR sequences (double underlined), the 218 linker (underlined and labeled) and selected restriction sites.
Figure 4 is a graphical representation of three competition ELISA's in which unlabeled SC-PEG unreacted CC49/218 SCA (closed squares), CC49/218 SCA (open squares), unlabeled XUS-PEG unreacted CC49/218 SCA (open circles), SC-PEG modified CC49/218 SCA (closed circles), XUS-PEG modified CC49/218 SCA (open triangles), CC49 IgG (closed triangles), an Anti-FITC SCA (dashed line) or BL-3 IgG (dotted line) were competed against a CC49 IgG radiolabeled with 125I for binding to the TAG- 72 antigen on a human breast carcinoma extract.
Figure 5 shows the pharmacokinetics of plasma retention of SCA and PEG-SCA. The details ofthe experiment are described in Example 13. Figure 6 shows an SDS-PAGE of the purified CC49-multimers cross- linked by PEG5000 under reducing conditions. The details ofthe experiment are described in Example 14. The lanes ofthe gel contain the following: 1) trimeric form; 2) dimeric form; 3) dimeric form; 4) mixed population; 5) native CC49; 6) PEG-CC49 monomer; 7) PEG-CC49 monomer; 8) empty; 9) empty; and 10) molecular weight standards.
Figure 7 shows the binding kinetics of Mono-, Di-, Tri-, -PEG-CC49. The details of the experiment are described in Example 14. Native CC49 is represented by the solid box. PEG-mono-CC49 is represented by the open box. PEG-Di-CC49 is represented by the solid diamond. PEG-Tri-CC49 is represented by the open diamond.
Figure 8 shows the results ofthe competition assay performed in Example 16. Nat is native CC49-SCA, C2 is PEG SC2000-CC49-SCA; GC is glyco- CC49-SCA; B* is the biotinylated CC49-SCA; C 12 is the PEG-SC 12,000-CC49- SCA; F5 is PEG-Flan-5000-CC49-SCA; and C20 is PEG-SC20000-CC49-SCA.
Description ofthe Embodiments
The present invention is directed to the novel combination of a polyalkylene glycol and a single chain polypeptide having binding affinity for an antigen, the polyalkylene glycol and polypeptide preferably being joined together by means of a coupling agent. Single Chain Polypeptides
The invention relates to the discovery that polyalkylene oxide conjugated single-chain antigen-binding proteins ("SCA") or single-chain variable fragments of antibodies ("sFv"), such as PEGylated SCA proteins, have significant utility beyond that of the nonPEGylated single-chain antigen-binding proteins. In addition to maintaining an antigen binding site, a PEGylated SCA protein has a PEG moiety which reduces antigenicity and increases the halflife ofthe modified polypeptide in the bloodstream. Accordingly, the invention is directed to monovalent and multivalent SCA proteins capable of PEGylation, compositions of monovalent and multivalent PEGylated SCA proteins, methods of making and purifying monovalent and multivalent PEGylated SCA proteins, and uses for PEGylated SCA proteins. The invention is also directed to PEGylated SCA proteins having a diagnostic or therapeutic agent covalently attached to an Cys- linked PEGylated polypeptide or an oligo-Lys linked PEGylated polypeptide. The terms "single-chain antigen-binding molecule" (SCA) or "single-chain
Fv" (sFv) are used interchangeably. They are structurally defined as comprising the binding portion of a first polypeptide from the variable region of an antibody VL (or VH), associated with the binding portion of a second polypeptide from the variable region of an antibody NH (or VL), the two polypeptides being joined by a peptide linker linking the first and second polypeptides into a single polypeptide chain, such that the first polypeptide is Ν-terminal to the linker and second polypeptide is C-terminal to the first polypeptide and linker. The single polypeptide chain thus comprises a pair of variable regions connected by a polypeptide linker. The regions may associate to form a functional antigen- binding site, as in the case wherein the regions comprise a light-chain and a heavy- chain variable region pair with appropriately paired complementarity determining regions (CDRs). In this case, the single-chain protein is referred to as a "single- chain antigen-binding protein" or "single-chain antigen-binding molecule." Single-chain Fvs can and have been constructed in several ways. Either VL is the N-terminal domain followed by the linker and VH (a VL -Linker- VH construction) or VH is the N-terminal domain followed by the linker and VL (VH -Linker- VL construction). The preferred embodiment contains VL in the N- terminal domain (see, Anand, N.N., et al, J. Biol Chem. 266:21874-21879 (1991)). Alternatively, multiple linkers have also been used. Several types of sFv (SCA) proteins have been successfully constructed and purified, and have shown binding affinities and specificities similar to the antibodies from which they were derived. A description ofthe theory and production of single-chain antigen-binding proteins is found in Ladner et al, U.S. Patent Nos. 4,946,778, 5,260,203, 5,455,030 and 5,518,889, and in Huston et al, U.S. Patent No. 5,091,513 ("biosynthetic antibody binding sites" (BABS)), all incoφorated herein by reference. The single-chain antigen-binding proteins produced under the process recited in the above patents have binding specificity and affinity substantially similar to that ofthe corresponding Fab fragment.
Typically, the Fv domains have been selected from the group of monoclonal antibodies known by their abbreviations in the literature as 26-10, MOPC 315, 741F8, 520C9, McPC 603, D1.3, murine phOx, human phOx, RFL3.8 sTCR, lA6, Sel55-4, 18-2-3, 4-4-20, 7A4-1, B6.2, CC49, 3C2, 2c, MA- 15C5/K12G0, Ox, etc. {see, Huston, J.S. et al, Proc. Natl. Acad. Sci. USA 55:5879-5883 (1988); Huston, J.S. et al, SIM News 38(4) (Supp.):\ \ (1988); McCartney, J. et al. CSU Short Reports 10:114 (1990); McCartney, J.E. et al, unpublished results (1990); Nedelman, M.A. et al, J. Nuclear Med. 32 (Supp.):\005 (1991); Huston, J.S. et al, In: Molecular Design and Modeling: Concepts and Applications, Part B, edited by J.J. Langone, Methods in Enzymology 203:46-88 (1991); Huston, J.S. et al, In: Advances in the Applications of Monoclonal Antibodies in Clinical Oncology, Epenetos, A.A. (Ed.), London, Chapman & Hall (1993); Bird, R.E. et al, Science 242:423-426 (1988); Bedzyk, W.D. et al, J. Biol. Chem. 265:18615-18620 (1990); Colcher, D. et al, J. Nat. Cancer Inst. 82:1191-1197 (1990); Gibbs, R.A. et al, Proc. Natl Acad. Sci. USA 88:4001 -4004 ( 1991 ); Milenic, D.E. et al. , Cancer Research 57:6363-6371 (1991); Pantoliano, M.W. et al, Biochemistry 30:10117-10125 (1991); Chaudhary, V.K. et al, Nature 339:394-397 (1989); Chaudhary, V.K. et al, Proc. Natl. Acad. Sci. USA 57:1066-1070 (1990); Batra, J.K. et al, Biochem. Biophys. Res. Comm. 77:1-6 (1990); Batra, J.K. etal., J. Biol Chem. 265:15198-15202 (1990); Chaudhary, V.K. et al, Proc. Natl. Acad. Sci. USA 57:9491-9494 (1990); Batra, J.K. et al, Mol. Cell Biol. 77:2200-2205 (1991); Brinkmann, U. et al, Proc. Natl. Acad. Sci. USA 55:8616-8620 (1991); Seetharam, S. et al, J. Biol. Chem. 266:1731 '6-17381 (1991); Brinkmann, U. et al, Proc. Natl. Acad. Sci. USA 59:3075-3079 (1992); Glockshuber, R. et al, Biochemistry 29:1362-1367 (1990); Skerra, A. et al, Bio/Technol 9:273-278 (1991); Pack, P. et al, Biochemistry 37:1579-1534 (1992); Clackson, T. et al, Nature 352:624-628 (1991); Marks, J.D. etal.,1 Mol. Biol. 222:581-597 (1991); Iverson, B.L. et al , Science 249:659-662 (1990); Roberts, V.A. et al , Proc. Natl. Acad. Sci. USA 57:6654-6658 (1990); Condra, J.H. et al, J. Biol. Chem. 265:2292-2295 (1990); Laroche, Y. et al, J. Biol. Chem. 266:16343-16349 ( 1991 ); Holvoet, P. et al. , J. Biol Chem. 266: 19717- 19724 ( 1991 ); Anand, N.N. et al, J. Biol. Chem. 266:21874-21879 (1991); Fuchs, P. et al, Bio/Technol. 9:1369-1372 (1991); Breitling, F. et al, Gene 104:104-153 (1991); Seehaus, T. et al, Gene 114:235-237 (1992); Takkinen, K. et al, Protein Engng. 4:837-841 (1991); Dreher, M.L. et al, J. Immunol. Methods 139:197-205 (1991); Mottez,
E. et al, Eur. J. Immunol. 27:467-471 (1991); Traunecker, A. et al, Proc. Natl. Acad. Sci. USA 55:8646-8650 (1991); Traunecker, A. et al, EMBO J. 10:3655- 3659 (1991); Hoo, W.F.S. et al, Proc. Natl. Acad. Sci. USA 59:4759-4763 (1993)).
Linkers of the invention used to construct sFv (SCA) polypeptides are designed to span the C-terminus of VL (or neighboring site thereof) and the N- terminus of VH (or neighboring site thereof) or between the C-terminus of VH and the N-terminus of VL. The preferred length ofthe peptide linker should be from 2 to about 50 amino acids. In each particular case, the preferred length will depend upon the nature ofthe polypeptides to be linked and the desired activity ofthe linked fusion polypeptide resulting from the linkage. Generally, the linker should be long enough to allow the resulting linked fusion polypeptide to properly fold into a conformation providing the desired biological activity. Where conformational information is available, as is the case with sFv (SCA) polypeptides discussed below, the appropriate linker length may be estimated by consideration ofthe 3 -dimensional conformation ofthe substituent polypeptides and the desired conformation ofthe resulting linked fusion polypeptide. Where such information is not available, the appropriate linker length may be empirically determined by testing a series of linked fusion polypeptides with linkers of varying lengths for the desired biological activity. Such linkers are described in detail in WO 94/12520, incoφorated herein by reference.
Preferred linkers used to construct sFv (SCA) polypeptides have between 10 and 30 amino acid residues. The linkers are designed to be flexible, and it is recommended that an underlying sequence of alternating Gly and Ser residues be used. To enhance the solubility ofthe linker and its associated single chain Fv protein, three charged residues may be included, two positively charged lysine residues (K) and one negatively charged glutamic acid residue (E). Preferably, one ofthe lysine residues is placed close to the N-terminus of VH, to replace the positive charge lost when forming the peptide bond ofthe linker and the VH.Such linkers are described in detail in U.S. Patent Application S. N. 08/224,591, filed April 7, 1994, incoφorated herein by reference. See also, Whitlow, M., et al, Protein Engng. 7:1017-1026 (1994). For multivalent sFvs (SCA), the association of two or more sFvs (SCA) is required for their formation. Although, multivalent sFvs (SCA) can be produced from sFvs (SCA) with linkers as long as 25 residues, they tend to be unstable. Holliger, P., et al, Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993), have recently demonstrated that linkers 0 to 15 residues in length facilitate the formation of divalent Fvs. See, Whitlow, M., et al. , Protein Engng. 7: 1017- 1026 (1994); Hoogenboom, H.R., Nature Biotech. 75:125-126 (1997); and WO 93/11161. a n object of the present invention is to produce a single-chain antigen- binding polypeptide-polyalkylene oxide conjugate which retains antigen binding affinity within a range of about two-fold to about ten-fold ofthe antigen binding affinity ofthe native single-chain antigen-binding polypeptide.
.Another object ofthe present invention is to produce an sFv (SCA) having one or more Cys residues such that the Cys residue is capable of being conjugated with PEG and the PEGylated polypeptide is capable of binding an antigen (i.e., the PEGylated polypeptide's ability to bind an antigen is not disrupted). A further object ofthe present invention is to produce an sFv (SCA) having three or more consecutive Lys residues such that the Lys residues are capable of being conjugated with PEG and the PEGylated polypeptide is capable of binding an antigen (i.e., the PEGylated polypeptide's ability to bind an antigen is not disrupted). These novel sFv (SCA) proteins may be conjugated to activated polyethylene glycol (PEG) such that the PEG modification occurs only (or preferentially) at the specifically engineered sites. The activated PEG molecules would be thiol-reactive or amine-reactive polymers such as are well known in the art. The designed changes correspond to amino acid residues on the sFv (SCA) surface which are well separated spatially from the antigen-binding site as deduced from known three-dimentional models ofthe antibody Fv domain.
A further object ofthe invention is to produce monovalent and multivalent sFvs (SCA) having one or more Cys PEG conjugation sequence(s). A further object of the invention is to produce monovalent and multivalent sFvs (SCA) having three or more consecutive Lys (i.e., oligo-Lys) PEG conjugation sequence(s). For multivalent sFv (SCA), the association of two or more sFvs (SC As) is required for their formation. For example, multivalent sFvs (SC As) may be generated by chemically crosslinking two sFvs (SCAs) with C-terminal cysteine residues (Cumber et al , J. Immunol. 749:120-126 (1992)) and by linking two sFvs (SCAs) with a third polypeptide linker to form a dimeric Fv (SCA)(George et al, J. Cell. Biochem. 15EΛ27 (1991)). Details for producing multivalent sFvs (SCAs) by aggregation are described in Whitlow, M., et al, Protein Engng. 7: 1017- 1026 ( 1994). Multivalent antigen-binding fusion proteins of the invention can be made by any process, but preferably according to the process formaking multivalent antigen-binding proteins set forth in WO 93/11161, incoφorated herein by reference.
Identification and Synthesis of Site Specific PEGylation Sequences
In the present invention, Cys PEGylation sites may occur in the VL and VH regions, adjacent to the C-terminus ofthe polypeptide (VL, VH or neighboring site thereof), the N-terminus ofthe polypeptide (VL, VH or neighboring site thereof), the linker region between the first and second polypeptide regions, or occur in a combination of these regions. In the present invention, oligo-Lys PEGylation sites may occur in the polypeptide linker or in the C-terminus or adjacent to the C-terminus of the polypeptide. The design of the PEG conjugaton sites on a protein involves examining the structural information known about the protein and the residues in the proteins involved in antigen binding. The PEG conjugaton sites are chosen to be as far from these residues as possible so as to prevent disruption ofthe antigen-binding site.
The Cys or the oligo-Lys PEGylation site may occur in (1) the native C- terminus of VL (or VH), (2) the C-terminus of VL (or VH) wherein the C-terminus has a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe peptide are sufficient for the PEGylated polypeptide to be capable of binding an antigen or (3) the C-terminus of VL (or VH) wherein the C-terminus has an addition of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe peptide are sufficient for the PEGylated polypeptide to be capable of binding an antigen. By "native" is intended the naturally occurring C-terminus ofthe immunoglobulin (first or second polypeptide). By "C-terminus" it is well understood in the art as intending the C-terminal amino acid residue or the C-terminal region of the polypeptide, which could include up to all of the amino acid residues of the polypeptide excluding the first N-terminal amino acid residue ofthe polypeptide. However, in the present invention, "C-terminus" is intended as the C-terminal amino acid residue ofthe above mentioned three types of C-terminus (1 , 2, or 3), unless otherwise indicated or intended.
PEGylation sites were identified and engineered at residues within loop sites in regions of the sFv (SCA) that are diametrically opposed to the antigen binding site. The five loop regions and C-terminal extension chosen as preferred sites of glycosylation are among the most distant regions spatially removed from the binding site.
The six furthest portions of an sFv (SCA) from the antigen binding site are as follows:
1) The loop made up of residues 11 to 15 in the light chain; 2) The loop made up of residues 77 to 79 in the light chain;
3) The N-terminus ofthe linker;
4) The loop made up of residues 11 to 15 in the heavy chain;
5) The loop made up of residues 82B, 82C and 83 in the heavy chain; and 6) The C-terminus ofthe sFv (or SCA).
The residues are identified as according to Kabat et al. , Sequences of Proteins of Immunological Interest, 5th ed., U.S. Dept. Health and Human Services, Bethesda, MD (1991). These possible PEGylation sites were determined by examining the 4-4-20 mouse Fab structure (see, Whitlow, M. et al, Protein Engng. 5:749-761 (1995), incoφorated herein by reference).
After identifying the loops furthest from the antigen binding site, the nucleic and amino acid sequences of each loop are examined for possible Cys PEGylation sites that may be engineered into the loop region. The engineered placement ofthe Cys residue anywhere in these six identified regions can generate a preferred site for sFv (SCA) PEGylation. The engineered placement of the oligo-Lys residues in the linker, the C-terminus ofthe sFv (SCA)andVor adjacent to the C-terminus of the sFv (SCA) can generate a preferred site for sFv PEGylation.
The design approach described above has been used for the CC49/218 SCA. Figure 2 shows the following resulting designs: designed PEGylation site no. 1 in the light chain ofthe CC49/218 SCA; designed PEGylation site no. 2 in the N-terminal end ofthe linker in CC49/218 SCA; designed PEGylation site no. 3 in the heavy chain ofthe CC49/218 SCA; designed PEGylation site no. 4 at the C-terminus of the CC49/218 SCA. Figure 3 shows the following resulting designs: designed oligo-Lys "hot spot" PEGylation sites at the C-terminus ofthe CC49/218 SCA. Any combination of these sites could be used.
The particular nucleotide sequence which is used to introduce a Cys or oligo-Lys PEGylation site into the various positions will depend upon the naturally-occurring nucleotide sequence. The most preferred sites are those in which it takes a minimum number of changes to generate the PEGylation site. Of course, based on the redundancy ofthe genetic code, a particular amino acid may be encoded by multiple nucleotide sequences.
Site-directed mutagenesis is used to change the native protein sequence to one that incoφorates the Cys residue or oligo-Lys residues for PEGylation. The mutant protein gene is placed in an expression system, such as bacterial cells, yeast or other fungal cells, insect cells or mammalian cells. The mutant protein can be purified by standard purification methods.
Oligonucleotide-directed mutagenesis methods for generating the Cys or oligo-Lys PEGylation sites and related techniques for mutagenesis of cloned DNA are well known in the art. See, Sambrook et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1989); Ausubel et al. (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley and Sons (1987), both incoφorated herein by reference. A preferred oligonucleotide-directed mutagenesis method for the present invention is according to Ho et al, Gene 77:51 -59 (1989), incoφorated herein by reference. Hosts and Vectors
After mutating the nucleotide sequence of the sFv (SCA), the mutated DNA can be inserted into a cloning vector for further analysis, such as for confirmation ofthe DNA sequence. To express the polypeptide encoded by the mutated DNA sequence, the DNA sequence is operably linked to regulatory sequences controlling transcriptional expression and introduced into either a prokaryotic or eukaryotic host cell.
Although sFvs (SCAs) are typically produced by prokaryotic host cells, eukaryotic host cells are the preferred host cells. Preferred host cells include yeast or other fungal cells, insect cells or mammalian cells. Standard protein purification methods may be used to purify these mutant proteins. Only minor modification to the native protein's purification scheme may be required.
Also provided by the invention are DNA molecules such as purified genetic sequences or plasmids or vectors encoding the sFv (SCA) ofthe invention that have engineered Cys residues and/or oligo-Lys residues capable of PEG conjugation. The DNA sequence for the PEGylated sFv (SCA) polypeptide can be chosen so as to optimize production in organisms such as prokaryotes, yeast or other fungal cells, insect cells or mammalian cells.
The DNA molecule encoding an sFv (SCA) having Cys residues and/or oligo-Lys residues for PEG conj ugation can be operably linked into an expression vector and introduced into a host cell to enable the expression ofthe engineered sFv (SCA) protein by that cell. A DNA sequence encoding an sFv (SCA) having Cys and/or oligo-Lys PEGylation sites may be recombined with vector DNA in accordance with conventional techniques. Recombina- nt hosts as well as methods of using them to produce single chain proteins ofthe invention are also provided herein.
The expression of such sFv (SCA) proteins of the invention can be accomplished in procaryotic cells. Preferred prokaryotic hosts include, but are not limited to, bacteria such as Neisseria, Mycobacte ia, Streptococci, Chlamydia and E. coli.
Eukaryotic hosts for cloning and expression of such sFv (SCA) proteins ofthe invention include insect cells, yeast, fungi, and mammalian cells (such as, for example, human or primate cells) either in vivo, or in tissue culture. A preferred host for the invention is Pichia pastor is.
The appropriate DNA molecules, hosts, methods of production, isolation and purification of monovalent, multivalent and fusion forms of proteins, especially sFv (SCA) polypeptides, are thoroughly described in the prior art, such as, e.g., U.S. Patent No. 4,946,778, which is fully incoφorated herein by reference.
The sFv (SCA) encoding sequence having Cys residues and/or oligo-Lys residues for PEG conjugation and an operably linked promoter may be introduced into a recipient prokaryotic or eukaryotic cell either as a non-replicating DNA (or RNA) molecule, which may either be a linear molecule or, more preferably, a closed covalent circular molecule. Since such molecules are incapable of autonomous replication, the expression of the desired sFv (SCA) protein may occur through the transient expression ofthe introduced sequence. Alternatively, permanent expression may occur through the integration of the introduced sFv (SCA) sequence into the host chromosome.
In one embodiment, the sFv (SCA) sequence can be integrated into the host cell chromosome. Cells which have stably integrated the introduced DNA into their chromosomes can be selected by also introducing one or more markers which allow for selection of host cells which contain the sFv (SCA) sequence and marker. The marker may complement an auxotrophy in the host (such as his4, leu2, or ura3, which are common yeast auxotrophic markers), biocide resistance, e.g., antibiotics, or resistance to heavy metals, such as copper, or the like. The selectable marker gene can either be directly linked to the sFv (SCA) DNA sequence to be expressed, or introduced into the same cell by co-transfection. In another embodiment, the introduced sequence will be incoφorated into a plasmid vector capable of autonomous replication in the recipient host cell. Any of a wide variety of vectors may be employed for this puφose. Factors of importance in selecting a particular plasmid or viral vector include: the ease with which recipient cells that contain the vector may be recognized and selected from those recipient cells which do not contain the vector; the number of copies ofthe vector which are desired in a particular host; and whether it is desirable to be able to "shuttle" the vector between host cells of different species.
Any of a series of yeast vector systems can be utilized. Examples of such expression vectors include the yeast 2-micron circle, the expression plasmids YEP 13, YCP and YRP, etc., or their derivatives. Such plasmids are well known in the art (Botstein et al, Miami Wntr. Symp. 19:265-274 (1982); Broach, J.R., In: The Molecular Biology of the Yeast Saccharomyces: Life Cycle and Inheritance, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, p. 445- 470 (1981); Broach, J.R., Cell 25:203-204 (1982)).
For a mammalian host, several possible vector systems are available for expression. One class of vectors utilize DNA elements which provide autonomously replicating extra-chromosomal plasmids, derived from animal viruses such as bovine papilloma viras, polyoma virus, adenovirus, or SV40 virus. A second class of vectors relies upon the integration ofthe desired gene sequences into the host chromosome. Cells which have stably integrated the introduced DNA into their chromosomes may be selected by also introducing one or more markers which allow selection of host cells which contain the expression vector. The marker may provide for prototrophy to an auxotrophic host, biocide resistance, e.g., antibiotics, or resistance to heavy metals, such as copper or the like. The selectable marker gene can either be directly linked to the DNA sequences to be expressed, or introduced into the same cell by co-transformation. Additional elements may also be needed for optimal synthesis of mRNA. These elements may include splice signals, as well as transcription promoters, enhancers, and termination signals. The cDNA expression vectors incoφorating such elements include those described by Okayama, H., Mol. Cell. Biol. 3:280 (1983), .and others.
.Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNHlόa, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Preferred vectors for expression in Pichia are pHIL-S 1 (Invitrogen Coφ.) and pPIC9 (Invitrogen Coφ.). Other suitable vectors will be readily apparent to the skilled artisan.
Once the vector or DNA sequence containing the constructs has been prepared for expression, the DNA constructs may be introduced or transformed into an appropriate host. Various techniques may be employed, such as transformation, transfection, protoplast fusion, calcium phosphate precipitation, electroporation, or other conventional techniques. After the cells have been transformed with the recombinant DNA (or RNA) molecule, the cells are grown in media and screened for appropriate activities. Expression of the sequence results in the production of the mutant sFv (SCA) for PEG conjugation of the present invention.
Straight chain Polymers
The straight chain polyalkylene gly cols employed in the practice of the present invention are ofthe structural formula
Figure imgf000033_0001
wherein R is selected from the group consisting of hydrogen, lower alkyl, and mixtures thereof, R1 is selected from the group consisting of hydrogen and lower alkyl, and n is a positive integer. By "lower alkyl" is meant an alkyl group having from one to four carbon atoms, i.e., methyl, ethyl, propyl, butyl, and isomers of the foregoing. R is preferably selected from the group consisting of hydrogen, methyl, and mixtures thereof, R1 is preferably selected from the group consisting of hydrogen and methyl, and n is preferably a positive integer of 500 or less. R is most preferably hydrogen, R1 is most preferably methyl, and n is most preferably an integer of 7 to 150. It will be readily apparent to those skilled in the art that the preferred poly(alkylene glycols) employed in the practice ofthe present invention are poly(ethylene glycol), poly(propylene glycol), mixtures thereof, and copolymers of poly(ethylene glycol) and poly(propylene glycol), wherein one of the terminal hydroxyl groups ofthe polymer may be substituted with a lower alkyl group. A preferred polyalkylene glycol for use in the present invention is poly(ethylene glycol)-hydrazine. The most preferred polyalkylene glycol for use in the present invention is methoxy poly(ethylene glycol). Hereinafter, for convenience, the polyalkylene glycol employed in the practice ofthe present invention will be designated PAG, which term is intended to include both compounds wherein R1 is hydrogen and compounds wherein R1 is alkyl. PEG refers to poly(ethylene glycol) and mPEG refers to methoxy poly(ethylene glycol). The PAG does not have to be of a particular molecular weight, but it is preferred that the molecular weight be between about 500 and about 40,000; more preferably, between about 2,000 and about 20,000. The choice of molecular weight of PAG is made based on the nature of the particular polypeptide employed, for example, the number of amino or other groups available on the polypeptide for modification. Molecular weights of about 10,000 and about 20,000 are most preferred.
It is well known in the art that PAGs that contain two terminal hydroxyl groups per moiety are capable of crosslinking other polymers, e.g. proteins. Where, as is often the case, crosslinking would be deemed undesirable, such crosslinking can be minimized or prevented by means known in the art. For example, Davis et al in U.S. Patent No. 4,179,337 have pointed out that a preferred means for preventing crosslinking is to preblock one end ofthe PAG, such as is done in the commercially available methoxy poly(ethylene glycol).
The P AGs employed in the practice ofthe present invention are preferably coupled to polypeptides by means of suitable coupling agents. A useful review of a number of coupling agents that can be employed in the practice ofthe present invention appears in Dreborg etal, Critical Reviews in Therapeutic Drug Carrier
Systems 6(^:315-365 (1990), see, especially, pp. 317-320.
Probably the best known coupling agent for this puφose is cyanuric chloride. Its use has been described in numerous references, see, for example, Abuchowski et al, J. Biol Chem. 252(17j:3578-3581 (June 10, 1977).
Zalipsky et al , Eur. Pol J. 19(12) : 1177- 1183 ( 1983), among others, have described the reaction of methoxy poly(ethylene glycol) with succinic .anhydride:
O
II mPEGOH + CH2 C O
\ O * mPEG— O— C II— CH,CH,COOH
/ CH2 C
It is also known to alkylate mPEG with ethylbromoacetate in the presence of a base such as K-tertiary butoxide in tertiary butanol, Na-naphthalene in tetrahydrofuran, or butyl lithium in benzene:
Base mPEGOH + BrCH2COOR *- mPEG — OCH2COOH
The terminal hydroxyl groups of PEG can be transformed into amine, carboxyl, or hexamethyl isocyanate groups. See, for example, Zalipsky et al, 1983, supra. A mixed anhydride derivative of carboxylated mPEG can be prepared in the presence of triethylamine and then reacted with proteins:
Figure imgf000036_0001
Figure imgf000036_0002
Carboxylated mPEG can also be reacted with hydroxysuccinimide in the presence of dicyclohexylcarbodumide and dimethyl formamide for reaction with protein:
Figure imgf000036_0003
King and Weiner {Int. J. Peptide Protein Res. 16:147 (1980) describe the dithiocarbonate of mPEG:
1. cs2 mPEGONa *> mPEG—OCSCH2CNH2
2. CICH9CONH9 II II
2 2 S O
Beauchamp et al, Analytical Biochem. 737:25-33 (1983) describe the activation of PEG with 1 , 1 '-carbonyldiimidazole. Reaction of this derivative with a peptide yields a carbamate linkage:
mPEGOH
Figure imgf000037_0001
Veronese et al, Appl. Biochem. & Biotechnol 77:141-152 (1985) describe the activation of methoxy poly(ethylene glycol) with phenylchloroformates, e.g., 2,4,5-trichlorophenylchloroformate or / nitrophenylchloroformate. These derivatives are linked to peptides by urethane linkages:
mPEGOH +
mPEGOH +
Figure imgf000037_0002
Ueno et al. in European Patent Application 87103259.5 form mPEG imidoesters from the corresponding nitriles by reaction with dry hydrogen chloride in the presence of a dehydrated lower alcohol:
mPEGOH + BrCH2CH2CH2CN- mPEGOCH2CH2CH2CN
CH3OH HCI
mPEGOCH2CH2CH2C----NH - HCI OCH3
NaH mPEGOH + CH2=CHCN * mPEG0CH2CH2CN
CH3OH HCI mPEG0CH2CH2C=NH HCI
I 0CH3
Abuchowski et al, Cancer Biochem. Biophys. 7.J75-186 (1984) have described forming mPEG succinate as described above and then forming methoxy polyethylene glycolyl succinimidyl succinate ("SS-PEG") by reaction with hydroxy succinimide in the presence of dicyclohexylcarbodumide:
mPEGOCCH2CH2COOH +
Figure imgf000038_0001
NH2-Protein mPEGOCCH2CH2C— NH-Protein
Figure imgf000038_0002
SS-PEG Sano et al, European Patent Application No. 89107960.0 disclose the phenyl glyoxal derivative of methoxy poly(ethylene glycol), which is capable of modifying the guanidino groups in peptides:
mPEGOH + Cl
mPEG
Figure imgf000039_0001
Zalipsky, in U.S. Patent No. 5,122,614, describes the activation of PEG by conversion into its N-succinimide carbonate derivative ("SC-PEG"): o o
II II mPEGOH + C—CI2 * mPEGOCCI
1. N-hydroxysuccinimide
2. triethylamine
Figure imgf000039_0002
met oxypoly(ethylene glycol) - succinyl carbonate SC - PEG
Zalipsky etal.,J. Macromol Sci. Chem. A2 :839, disclose the .amino acid ester derivative of methoxy poly(ethylene glycol):
O R
II I
PEG— O— C— CH— H2 Davis et al. , U.S. Patent No.4, 179,337, disclose a hydrazide derivative of methoxy poly(ethylene glycol), which is capable of modifying aldehydes and ketones and other functional groups:
0
II
PEG— O — CH2 — C— NH — NH3 +Cr HZ-PEG
It is further disclosed that the bifunctional derivative of PEG, i.e., polyethylene glycol-bis-succinidyl carbonate ("BSC-PEG") can be prepared by similar means. The SC-PEG and BSC-PEG compounds are then reacted with amine groups in a protein and attached thereto via urethane (carbamate) linkages.
It will be readily apparent to those skilled in the art that other activated PAGs can also be employed in the practice ofthe present invention. The preferred activated PAG for use in the practice ofthe present invention is selected from the group consisting of SS-PEG and SC-PEG. The use of SC-PEG is most preferred.
Branched Polymers
The invention further provides for the use of branched, substantially non- antigenic polymers for polyalkylene oxide conjugation ofthe sFv (SCA) proteins corresponding to the formula:
(R)nL-A (II) wherein (R) includes a water-soluble non-antigenic polymer; (n) = 2 or 3; (L) is an aliphatic linking moiety covalently linked to each (R); and
(A) represents an activated functional group capable of undergoing nucleophilic substitution. For example, (A) can be a group which is capable of bonding with biologically active nucleophiles or moieties capable of doing the same.
In particularly preferred aspects of the invention (R) includes a poly(alkylene oxide) PAO such as poly(ethylene glycol) PEG or mPEG. It is preferred that each chain have a molecular weight of between about 200 and about 12,000 daltons and preferably between about 1,000 and about 10,000 daltons. Molecular weights of about 5,000 daltons are most preferred.
As shown in Formula II, 2 or 3 polymer chains, designated (R) herein, are joined to the alphatic linking moiety (L). Suitable aliphatics included substituted alkyl diamines and triamines, lysine esters and malonic ester derivatives. The linking moieties are perferably non-planar, so that the polymer chains are not rigidly fixed. The linking moiety (L) is also a means for attaching the mulitple polymer chains or "branches" to (A), the moeity through which the polymer attaches to the sFv (SCA) protein.
(L) preferably includes a multiply-functionalized alykyl group containing up to 18, and more preferably between 1-10 carbon atoms. A heteroatom such as nitrogen, oxygen or sulfur may be included within the alkyl chain. The alkyl chain may also be branched at a carbon or nitrogen atom. In another aspect ofthe invention, (L) is a single nitrogen atom.
(L) and (R) are preferably joined by a reaction between nucleophilic functional groups on both (R) and (L). Each (R) is suitably functionalized to undergo nucleophilic substitution and bond with (L). Such functionalization of polymers is readily apparent to those of ordinary skill in the art. A wide variety of linkages are contemplated between (R) and (L).
Urethane (carbamate) linkages are preferred. The bond can be formed, for example, by reacting an amino group such as l,3-diamino-2-propanol with methoxypolyethylene glycol succinimidyl carbonate as described in U.S. Patent No. 5,122,614. -Amide linkages, which can be formed by reacting an amino- terminated non-antigenic polymer suchas methoxypolyethylene glycol-amine (mPEG amine) with an acyl chloride functional group. Examples of other such linkages include ether, amine, urea, and thio and thiol analogs thereof, as well as the thio and thiol analogs ofthe urethane and amide linkages discussed supra. The moiety (A) of Formula II represents groups that "activate" the branched polymers ofthe present invention for conjugation with biologically active materials. (A) can be a moiety selected from:
1. Functional groups capable of reacting with an amino group such as: a) carbonates such as the p-nitrophenyl or succinimidyl; b) carbonyl imidazole; c) azlactones; d) cyclic imide thiones; or e) isocyanates or isothiocyanates. 2. Functional groups capable of reacting with carboxylic acid groups and reactive with carbonyl groups such as: a) primary amines; or b) hydrazine and hydrazide functional groups such as the acyl hydrazides, carbazates, semicarbamates, thiocarbazates, etc. 3. Functional groups capable of reacting with mercapto or sulfhydryl groups such as phenyl glyoxals; see, for example, U.S. Patent No. 5,093,531.
4. Other nucleophiles capable of reacting with an electrophilic center. A non-limiting list includes, for example, hydroxyl, amino, carboxyl, thiol groups, active methylene and the like. The moiety (A) can also include a spacer moiety located proximal to the aliphatic linking moiety (L). The spacer moiety may be a heteroalkyl, alkoxyl, alkyl containing up to 18 carbon atoms or even an additional polymer chain. The spacer moieties can be added using standard synthesis techniques.
The branched polymers, generally, U-PAO's or U-PEG's, are formed using conventional reaction techniques known to those of ordinary skill in the art.
These umbrella-like branched polymers ofthe present invention (U-PAO' s or U-PEG's) react with biologically active nucleophiles to form conjugates. The point of polymer attachment depends upon the functional group (A). For example,
(A) can be a succinimidyl succinate or carbonate and react with e-amino lysines. The branched polymers can also be activated to link with any primary or secondary amino group, mercapto group, carboxylic acid group, reactive carbonyl group or the like found on biologically active polypeptides. Other groups are apparent to those of ordinary skill in the art.
One ofthe main advantages ofthe use ofthe branched polymers is that the branching imparts an umbrella-like three dimensional protective covering to the materials they are conjugated with. This contrasts with the string-like structure ofthe straight chain polymers discussed, supra. .An additional advantage of the branched ploymers is that they provide the benefits associated with attaching several strands of polymers to a sFv protein but require substantially fewer conjugation sites. The desired properties of PEGylation are realized and the loss of bioactivity is minimized.
One or more of the activated branched polymers can be attached to a biologically active nucleophile, such as an sFv protein, by standard chemical reactions. The conjugate is represented by the formula:
[(R^L-A'k - (nucleophile) (III)
wherein (R) is a water-soluble substantially non-antigenic polymer; n=2 or 3; (L) is an aliphatic linking moiety; (A1) represents a linkage between (L) and the nucleophile and (z) is an integer > 1 representing the number of polymers conjugated to the biologically active nucleophile. The upper limit for (z) will be determined by the number of available nucleophilic attachment sites and the degree of polymer attachment sought by the artisan. The degree of conjugation can be modified by varying the reaction stoichimetry using well-known techniques. More than one polymer conjugated to the nucleophile can be obtained by reacting a stoichimetric excess ofthe activated polymer with the nucleophile. Purification ofsFv Proteins
A generic protocol that has been developed and used to produce twelve different single chain antigen binding molecules. It involves cell lysis and washing, solubilization in a denaturing solvent, refolding by dilution, and two ion-exchange HPLC chromatography steps. Such isolated sFvs (SCAs) are capable of being
PAG conjugated according to the present invention.
The fermentation of the sFv-producing E. coli strains are performed at 32 °C using a casein digest-glucose-salts medium. At an optical density of 18 to 20 at 600 nm, sFv expression is induced by a 42° C temperature shock for one hour. After the fermentation is cooled to 10°C, the cells are harvested by centrifugation at 7000g for ten minutes. The wet cell paste is then stored frozen at -20 °C. Approximately 200 to 300 g of wet cell paste is normally recovered from one 10-liter fermentation.
For protein recovery, the cell paste from three 10-liter fermentations (600- 900 g) is thawed overnight at 4 °C and gently resuspended at 4 °C in 50 mM Tris- HC1, 1.0 mM EDTA, 100 mM KC1, 0J mM phenylmethylsulfonyl chloride (PMSF), pH 8.0 (lysis buffer), using 10 liters of lysis buffer for every kilogram of wet cell paste. When thoroughly resuspended, the chilled mixture is passed three times through a Manton-Gaulin cell homogenizer to fully lyse the cells. Because the cell homogenizer raises the temperature ofthe cell lysate to 25±5 °C, the cell lysate is cooled to 5±2°C with a Lauda/Brinkman chilling coil after each pass. Complete lysis is verified by visual inspection under a microscope.
The cell lysate is centrifuged at 24,3 OOg for thirty minutes at 6°C using a Sorvall RC-5B centrifuge. The pellet contains the insoluble sFv and the supernatant is discarded. The pellet is washed by gently scraping it from the centrifuge bottles and resuspending it in 5 liters of lysis buffer/kg of wet cell paste. The resulting 3.0-4.5-liter suspension is again centrifuged at 24,300g for 30 min at 6°C, and the supernatant is discarded. This washing ofthe cell pellet removes soluble E. coli proteins and can be repeated as many as five times. At any time during this washing procedure the material can be stored as a frozen pellet at -20°C. A substantial time saving in the washing steps can be accomplished by utilizing a Pellicon tangential flow apparatus equipped with 0.22-μm microporous filters. The washed cell pellet is solubilized at 4°C in freshly prepared 6 M guanidine hydrochloride, 50 mM Tris-HCl, 10 mM CaCl2, 50 mM KC1, pH 8.0 (denaturing buffer), using 6 ml/g of pellet. If necessary, a few quick pulses from a Heat Systems Ultrasonics tissue homogenizer can be used to complete the solubilization. The resulting suspension is centrifuged at 24,300g for 45 minutes at 6°C and the pellet is discarded. The optical density of the supernatant is determined at 280 nm and if the OD2g0 is above 30, additional denaturing buffer is added to obtain an OD280 of approximately 25.
The supernatant is slowly diluted into cold (4-7 °C) refolding buffer (50 mM Tris-HCl, 10 mM CaCl2, 50 mM KC1, 0J mM PMSF, pH 8.0) until a 1 :10 to 1 :100 dilution is reached (final volume 70-120 liters). The refolding buffer should be prepared at least one day prior to use, to allow sufficient time for it to cool to 4°C. The best results will be obtained when the supernatant is slowly added to the refolding buffer over a two hour period, with gentle mixing. The solution is left undisturbed for at least twenty hours and then filtered through a Millipore Pellicon tangential flow apparatus at 4°C with four to six 0.45-μm microporous membranes (HVLP 000 C5). The filtrate is concentrated to 1 to 2 liters using a Pellicon apparatus with four to six 10,000 NMWL cassettes (SK1PA156A4), again at 4°C.
The concentrated crude sFv sample is buffer exchanged at 4°C into 20 mM 2-[N-moφholino]ethanesulfonic acid (Mes), 0.3 mM CaCl2, pH 6.0, using the Pellicon ultrafiltration apparatus equipped with four to six 10,000 NMWL cassettes. The sample is then chromatographed on a Waters Accell Plus CM ion- exchange (RCM) column (4.7 x 30.0 cm). Prior to loading on the HPLC, the material is filtered through a 0.22-μm filter and the Accell column is equilibrated with Buffer A (40 mM Mes, 1 mM CaCl2, pH 6.0). Following sample loading, the Accell column is eluted over a 55 -minute period with a linear gradient of Buffer A and Buffer B (40 mM Mes, 100 mM CaCl2, pH 7.0). (See Table 1).
Figure imgf000046_0001
The Accell Plus CM column has a capacity of about 3 g and thus all the crude sFv sample can normally be loaded in a single run. The fractions are analyzed using 4-20% Novex SDS-PAGE gels and the peak fractions are pooled.
Normally, the sFv elutes from the Accell ion-exchange column quite early in the gradient. To enhance resolution for certain sFv proteins, holds in the gradient can be implemented. The pooled fractions from the Accell HPLC purification are dialyzed against Buffer D (40 mM 3-[N-moφholino]propanesulfonic acid (Mops), 0.5 mM
Ca acetate, pH 6.0) until the conductivity is lowered to that of Buffer D. The sample is then loaded on a 21.5 x 150-mm poly aspartic acid Poly CAT A column.
If more than 60 mg is loaded on this column, the resolution begins to deteriorate; thus, the pooled fractions from the Accell HPLC purification often must be divided into several PolyCAT A runs. Most sFv proteins have an extinction coefficient of about 2.0 mg ml"1 cm"1 at 280 nm and this can be used to determine protein concentration. The sFv sample is eluted from the PolyCAT A column with a 50-min linear gradient of Buffer D and Buffer E (40 mM Mops, 10 mM Ca acetate, pH 8.0). See Table 2.
Figure imgf000047_0001
The sFv proteins will often elute between 20 and 26 min when this gradient is used. This corresponds to an eluting solvent composition of approximately 70% Buffer D and 30% Buffer E.
This purification procedure yields sFv proteins that are more than 95% pure as examined by SDS-PAGE and Scatchard analysis. Modifications ofthe above procedure may be dictated by the isoelectric point of the particular sFv being purified, which is often between 8.0 and 9.3.
The polyalkylene gly cols (PAGs) employed in the practice ofthe present invention, which, as indicated above, are preferably activated by reaction with a coupler, can be reacted with any of several groups that may be present attached to the chain ofthe single chain antigen binding molecules, e.g. terminal carboxyl groups, thiol groups, phenolic hydroxyl groups, or primary amino groups located at the chain terminus or along the chain. It is preferred to react activated PAGs with primary amine groups, especially those occurring along the peptide chain. It is most preferred that the activated PAGs be coupled to the e amino groups of lysine residues as well as cysteine residues in the polypeptide.
The reaction between the PAG and the single chain polypeptide is normally carried out in solution, preferably an aqueous buffer solution providing a pH in the range of from about 6 to about 10, preferably from about 7 to about 9, most preferably from about 7 to about 8. As examples of buffer solutions that will provide pH's in these ranges at 25° C may be listed:
50 ml of 0 J molar potassium dihy drogen phosphate + 5.6 to 46 J ml OJ molar NaOH diluted to 100 ml 50 ml of 0.025 molar borate + 2.0 to 20.5 ml 0J molar HCI diluted to 100 ml 50 ml of 0.025 molar borate + 0.9 to 18.3 ml 0J molar NaOH diluted to 100 ml 50 ml of 0.05 molar sodium bicarbonate + 5.0 to 10.7 ml 0J molar NaOH diluted to 100 ml
The precise adjustment of the quantity of acid or base to be used to provide a particular desired pH will be readily determinable by those skilled in the art.
If, in a given instance, the use of a biological buffer should be required, one ofthe following may be employed:
3-(N-Moφholino)propanesulfonic acid (MOPS)
3-(N-Mθφholino)-2-hydroxypropanesufonic acid (MOPSO) Piperazine-N,N'-bis(2-hydroxypropanesulfonic acid) (POPSO)
The reaction between the PAG and the single chain polypeptide will normally be run under conditions that will not give rise to denaturation, e.g. mild temperatures and no more agitation than necessary. The reaction will preferably be run at a temperature in the range of from about 4° C to about 25° C. More preferably, the reaction will be run at room temperature, i.e. from about 20° C to about 25° C.
It will be readily understood by those skilled in the art that the amount of PAG employed relative to the amount of single chain polypeptide will be dependent upon the desired nature ofthe reaction product. Where, for example, it is desired to react a PAG with each lysine residue along the polypeptide chain, an amount of PAG at least equimolar to the lysine concentration will be required. It will be advantageous to employ an excess of PAG, where possible, in order to increase the reaction rate and the likelihood of a complete reaction. Clearly, if fewer than all ofthe possible reaction sites along the polypeptide chain are to be derivatized, correspondingly less PAG will be used. In general, however, where molar excesses of PAG's are used, it has been determined that molar excesses on the order of 2 - 100 of the PAG can be used; molar excesses of 2 - 10 are preferred. The time required for the reaction will depend upon a number of factors, such as reaction temperature, concentration of reactants, and whether full or partial reaction is desired. The course of the reaction can be monitored by conventional means, such as the analysis of periodic samples by size exclusion chromatography or gel electrophoresis. The reaction can conveniently be terminated when desired by the addition of a compound having a primary amine group, e.g. glycine, to scavenge the excess PAG. A reaction time of about 15 - 120 minutes will typically be required to fully react the PAG with the primary amine groups of the lysine residues of the single chain polypeptide at room temperature. The skilled practitioner will understand that the time for conjugation, as well as the amount and type of PAG, must not be such as to inactivate the polypeptide being employed.
Purification ofthe PAG/single chain polypeptide reaction product can be effected by means commonly employed by those skilled in the art, such as, for example, size exclusion chromatography, ion-exchange chromatography, ultrafiltration, dialysis, and the like. Solutions of the reaction product can, if desired, be concentrated with a rotary evaporator and can be obtained in the dry state by lyophilization.
Depending upon the particular single chain antigen binding molecule chosen and the extent to which it is reacted with the PAG, the resulting adduct is expected to be useful both diagnostically and therapeutically, exhibiting, as compared to the unreacted single chain polypeptide, decreased immunogenicity, increased circulating life, and increased stability while maintaining an acceptable level of activity.
The single chain antigen binding polypeptide can be reacted with the activated branched polyethylene glycol polymers discussed above in an aqueous reaction medium which can be buffered, depending on the pH requirements ofthe nucleophile. The optimum pH for the reaction is generally between about 6.5 and about 8.0 and preferably about 7.4 for polypeptides. The optimum reaction conditions for the sFv stability, reaction efficiency, etc., is within the level of ordinary skill in the art. The preferred temperature range is between 4°C and 37 °C. The reaction temperature cannot exceed the temperature at which the nucleophile may denature or decompose. It is preferred that the nucleophile be reacted with an excess ofthe activated branched polymer. Following the reaction, the cojugate is recovered and purified, for example, by diafiltration, column chromatography, combinations thereof, or the like.
Conjugates
Upon production ofthe polyalkylene oxide conjugated sFv (SCA) ofthe present invention, the polyalkylene oxide conjugated sFv may further be modified by conjugating a diagnostic or therapeutic agent to the polyalkylene oxide conjugated sFv. The general method of preparing an antibody conjugate according to the invention is described in Shih, L.B., etal, Cancer Res. 57:4192 (1991); Shih, L.B., andD.M. Goldenberg, Cancer Immunol. Immunother. 31:197 (1990); Shih, L.B., et al, Intl. J. Cancer 46:1101 (1990); Shih, L.B., et al, Intl. J. Cancer 41:832 (1988), all incoφorated herein by reference. The indirect method involves reacting an antibody (or sFv), whose polyalkylene oxide has a functional group, with a carrier polymer loaded with one or plurality of bioactive molecules, such as, peptides, lipids, nucleic acids (i.e., phosphate-lysine complexes), drug, toxin, chelator, boron addend or detectable label molecule(s). Alternatively, the polyalkylene oxide conjugated sFv may be directly conjugated with a diagnostic or therapeutic agent. The general procedure is analogous to the indirect method of conjugation except that a diagnostic or therapeutic agent is directly attached to an oxidized sFv component. See Hansen et al, U.S. Patent No. 5,443,953, incoφorated herein by reference.
The polyalkylene oxide conjugated sFv can be attached to a derivative of the particular drug, toxin, chelator, boron addend or label to be loaded, in an activated form, preferably a carboxyl-activated derivative, prepared by conventional means, e.g., using dicyclohexylcarbodumide (DCC) or a water soluble variant thereof, to form an intermediate adduct.
Many drugs and toxins are .known which have a cytotoxic effect on tumor cells or microorganisms that may infect a human and cause a lesion, in addition to the specific illustrations given above. They are to be found in compendia of drugs and toxins, such as the Merck Index and the like. .Any such drug can be loaded onto a carrier or directly onto a polyalkylene oxide conjugated sFv by conventional means well known in the art, and illustrated by analogy to those described above.
Chelators for radiometals or magnetic resonance enhancers are also well known in the art. Typical are derivatives of ethylenediaminetetraacetic acid (EDTA) and diethylenetriaminepentaacetic acid (DTP A). These typically have groups on the side chain by which the chelator can be attached to a carrier or directly onto a polyalkylene oxide conjugated sFv. Such groups include, e.g., a benzylisothiocyanate, by which the DTP A or EDTA can be coupled to the reactive group of an sFv. Labels such as radioisotopes, enzymes, fluorescent compounds, electron transfer agents, .and the like can also be linked to carrier or directly onto a polyalkylene oxide conjugated sFv by conventional methods well known to the art. These labels and the sFv conjugates prepared from them can be used for immunoassays and for immunohistology, much as the sFv conjugate prepared by direct attachment ofthe labels to the sFv. However, the loading ofthe conjugates according to the present invention with a plurality of labels can increase the sensitivity of assays or histological procedures, where only low extent of binding ofthe sFv to target antigen is achieved. Boron addends, e.g., carboranes, when attached to single-chain antigen binding molecules and targeted to lesions, can be activated by thermal neutron irradiation and converted to radioactive atoms which decay by alpha emission to produce highly cytotoxic short-range effects. High loading of boron addends, as well as of magnetic resonance enhancing ions, is of great importance in potentiating their effects. Carboranes can be made with carboxyl functions on pendant side chains, as is well known in the art.
Loading of drugs on the carrier will depend upon the potency ofthe drug, the efficiency of sFv targeting and the efficacy ofthe conjugate once it reaches its target. In most cases, it is desirable to load at least 20, preferably 50, and often 100 or more molecules of a drug on a carrier. The ability to partially or completely detoxify a drug as a conjugate according to the invention, while it is in circulation, can reduce systemic side effects ofthe drug and permit its use when systemic administration of the unconjugated drug would be unacceptable. Administration of more molecules of the drug, but conjugated to the sFv on a carrier, according to the present invention, permits therapy while mitigating systemic toxicity.
Toxins will often be less heavily loaded than drugs, but it will still be advantageous to load at least 5, preferably 10 and in some cases 20 or more molecules of toxin on a carrier and load at least one carrier chain on the sFv for targeted delivery. Uses
The polyalkylene oxide conjugated sFv (SCA) polypeptide conjugates of the present invention are expected to have much longer circulating half lifes and reduced immunogenicity in vivo. This may solve a potential limitation relating to very rapid blood clearance of some sFv proteins. It would also reduce or eliminate concerns about repeated administration of a therapeutic sFv which may otherwise provoke an immune response in the patient. The choice of the particular cysteine and/or oligo-lysine mutant combinations may allow one to achieve circulating lives over a considerable range depending on the specific polyalkylene oxide conjugated sFv variant polypeptide. This would allow sFv to be administered for the therapeutic use of choice.
A diagnostic or therapeutic agent is a molecule or atom which is conjugated to an antibody and useful for diagnosis or for therapy. The immunoreactivity ofthe antibody is retained. Diagnostic or therapeutic agents include drugs, toxins, chelators, boron compounds and detectable labels. See "Conjugates" section, supra, for further details.
The diagnostic or therapeutic agent may be, but is not limited to, at least one selected from a nucleic acid, a compound, a protein, an element, a lipid, an antibody, a saccharide, an isotope, a carbohydrate, an imaging agent, a lipoprotein, a glycoprotein, an enzyme, a detectable probe, or any combination thereof, which may be detectably labeled as for labeling antibodies, as described herein. Such labels include, but are not limited to, enzymatic labels, radioisotope or radioactive compounds or elements, fluorescent compounds or metals, chemiluminescent compounds and bioluminescent compounds. Alternatively, any other known diagnostic or therapeutic agent can be used in a method ofthe present invention.
A therapeutic agent used in the present invention may have a therapeutic effect on the target cell, the effect selected from, but not limited to, correcting a defective gene or protein, a drug action, a toxic effect, a growth stimulating effect, a growth inhibiting effect, a metabolic effect, a catabolic affect, an anabolic effect, an antiviral effect, an antibacterial effect, a hormonal effect, a neurohumoral effect, a cell differentiation stimulatory effect, a cell differentiation inhibitory effect, a neuromodulatory effect, an antineoplastic effect, an anti-tumor effect, an insulin stimulating or inhibiting effect, a bone marrow stimulating effect, a pluripotent stem cell stimulating effect, an immune system stimulating effect, and any other known therapeutic effects that may be provided by a therapeutic agent delivered to a cell via a delivery system according to the present invention.
The sFv conjugate of the present invention may be used for protection, suppression or treatment of infection or disease. By the term "protection" from infection or disease as used herein is intended "prevention," "suppression" or "treatment." "Prevention" involves administration of a glycosylated sFv conjugate prior to the induction ofthe disease. "Suppression" involves administration ofthe composition prior to the clinical appearance ofthe disease.
"Treatment" involves administration ofthe protective composition after the appearance of the disease. It will be understood that in human and veterinary medicine, it is not always possible to distinguish between "preventing" and "suppressing" since the ultimate inductive event or events may be unknown, latent, or the patient is not ascertained until well after the occurrence of the event or events. Therefore, it is common to use the term "prophylaxis" as distinct from "treatment" to encompass both "preventing" and "suppressing" as defined herein. The term "protection," as used herein, is meant to include "prophylaxis."
Such additional therapeutic agents which can further comprise a therapeutic agent or composition ofthe present invention may be selected from, but are not limited to, known and new compounds and compositions including antibiotics, steroids, cytotoxic agents, vasoactive drugs, antibodies and other therapeutic modalities. Non-limiting examples of such agents include antibiotics used in the treatment of bacterial shock, such as gentamycin, tobramycin, nafcillin, parenteral cephalosporins, etc; adrenal corticosteroids and analogs thereof, such as methyl prednisolone, mitigate the cellular injury caused by endotoxins; vasoactive drugs, such as alpha receptor blocking agent (e.g., phenoxybenzamine), beta receptor agonists (e.g., isoproterenol), and dopamine are agents suitable for treating septic shock.
Polyalkylene oxide conjugated sFv ofthe invention may also be used for diagnosis of disease and to monitor therapeutic response. Other uses of polyalkylene oxide conjugated sFv proteins are specific targeting of pro-drug activating enzymes to tumor cells by a bispecific molecule with specificity for tumor cells and enzyme. Polyalkylene oxide conjugated sFv may be used for specific delivery of drug to an in vivo target, such as a tumor, delivery of radioactive metals for tumor radioimmunodiagnosis or radioimmunotherapy (Goldenberg, D.M., Am. J. Med. 94:297 (1993)), nonradioactive metals in applications such as with boron/uranium-neutron capture therapy (Ranadive, G.N., et al, Nucl. Med. Biol. 20:1 (1993); Barth, R.F., et al, Bioconjug. Chem. 5:58 (1994)), and nuclear magnetic resonance imaging (Sieving, P.F., etal, Bioconjug. Chem. 7:65 (1990)). This list is illustrative only. The invention also extends to uses for the polyalkylene oxide conjugated sFv proteins in purification and biosensors. Affinity purification is made possible by affixing the polyalkylene oxide conjugated sFv protein to a support, with the antigen-binding sites exposed to and in contact with the ligand molecule to be separated, and thus purified. Biosensors generate a detectable signal upon binding of a specific antigen to an antigen-binding molecule, with subsequent processing of the signal. Polyalkylene oxide conjugated sFv proteins, when used as the antigen-binding molecule in biosensors, may change conformation upon binding, thus generating a signal that may be detected.
The invention is also directed to a method of detecting an antigen suspected of being in a sample by contacting the sample with the polyalkylene oxide conjugated sFv that is labeled. A sample may comprise at least one compound, mixture, surface, solution, emulsion, suspension, mixture, cell culture, fermentation culture, cell, tissue, secretion and/or derivative or extract thereof.
Such samples can also include, e.g., animal tissues, such as blood, lymph, cerebrospinal fluid (CNS), bone marrow, gastrointestinal contents, and portions, cells or internal and external secretions of skin, heart, lung and respiratory system, liver, spleen, kidney, pancreas, gall bladder, gastrointestinal tract, smooth, skeletal or cardiac muscle, circulatory system, reproductive organs, auditory system, the autonomic and central nervous system, and extracts or cell cultures thereof. Such samples can be measured using methods ofthe present invention in vitro, in vivo and in situ.
Such samples can also include environmental samples such as earth, air or water samples, as well as industrial or commercial samples such as compounds, mixtures, surfaces, aqueous chemical solutions, emulsions, suspensions or mixtures.
Additionally, samples that can be used in methods ofthe present invention include cell culture and fermentation media used for growth of prokaryotic or eukaryotic cells and/or tissues, such as bacteria, yeast, mammalian cells, plant cells and insect cells. Essentially all ofthe uses for which monoclonal or polyclonal antibodies, or fragments thereof, have been envisioned by the prior art, can be addressed by the polyalkylene oxide conjugated sFv proteins ofthe present invention. These uses include detectably-labeled forms ofthe polyalkylene oxide conjugated sFv protein. Types of labels are well-known to those of ordinary skill in the art. They include radiolabeling, chemiluminescent labeling, fluorochromic labeling, and chromophoric labeling. Other uses include imaging the internal structure of an animal (including a human) by administering an effective amount of a labeled form of the polyalkylene oxide conjugated sFv protein and measuring detectable radiation associated with the animal. They also include improved immunoassays, including sandwich immunoassay, competitive immunoassay, and other immunoassays wherein the labeled antibody can be replaced by the PEGylated sFv protein of this invention. See, e.g., Kohler etal, Nature 256:495 (1975); Kohler et al, Eur. J. Immunol. 6:511 (1976); Kohler et al, Eur. J. Immunol. 6:292 { 1976); Hammerling et al , In: Monoclonal Antibodies and T-Cell Hybridomas, pp. 563-681, Elsevier, N (1981); Sambrook et al, Molecular Cloning - A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory (1989).
Administration
Administration of polyalkylene oxide conjugated sFv conjugates of the invention for in vivo diagnostic and therapeutic applications will be by analogous methods to sFv where the diagnostic or therapeutic principle is directly linked to the sFv or a loaded carrier is linked by random binding to amine or carboxyl groups on amino acid residues ofthe sFv in a non-site-specific manner.
Conjugates ofthe present invention (immunoconjugates) can be formulated according to known methods to prepare pharmaceutically useful compositions, such as by admixture with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their formulation are described, for example, in Remington 's Pharmaceutical Sciences, 18th ed., Osol, A., ed., Mack, Easton PA (1990). In order to form a pharmaceutically acceptable composition suitable for effective administration, such compositions will contain a therapeutically effective amount ofthe immunoconjugate, either alone, or with a suitable amount of carrier vehicle.
Additional pharmaceutical methods may be employed to control the duration of action. Controlled release preparations may be achieved by the use of polymers to complex or absorb the immunoconjugate ofthe present invention. The controlled delivery may be exercised by selecting appropriate macromolecules (for example, polyesters, polyamino acids, polyvinyl pyrrolidone, ethylene- vinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate). The rate of drug release may also be controlled by altering the concentration of such macromolecules. Another possible method for controlling the duration of action comprises incoφorating the therapeutic agents into particles of a polymeric substance such as polyesters, polyamino acids, hydrogels, poly(lactic acid) or ethylene vinylacetate copolymers. Alternatively, it is possible to entrap the immunoconjugate ofthe invention in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, by the use of hydroxymethylcellulose or gelatin-microcapsules or poly(methylmethacrylate) microcapsules, respectively, or in a colloid drug delivery system, for example, liposomes, albumin microspheres, microemulsions, nanoparticles, nanocapsules, or in macroemulsions. Such teachings are disclosed in Remington's Pharmaceutical Sciences, 16th ed., Osol, A., ed., Mack, Easton PA (1990).
The immunoconjugate may be provided to a patient by means well known in the art. Such means of introduction include oral means, intranasal means, subcutaneous means, intramuscular means, intravenous means, intra-arterial means, or parenteral means. Intravenous, intraarterial or intrapleural administration is normally used for lung, breast, and leukemic tumors. Intraperitoneal administration is advised for ovarian tumors. Intrathecal administration is advised for brain tumors and leukemia. Subcutaneous administration is advised for Hodgkin's disease, lymphoma and breast carcinoma. Catheter perfusion is useful for metastatic lung, breast or germ cell carcinomas of the liver. Intralesional administration is useful for lung and breast lesions.
For therapeutic or diagnostic applications, compositions according to the invention may be administered parenterally in combination with conventional injectable liquid carriers such as sterile pyrogen-free water, sterile peroxide-free ethyl oleate, dehydrated alcohol, or propylene glycol. Conventional pharmaceutical adjuvants for injection solution such as stabilizing agent, solubilizing agents and buffers, such as ethanol, complex forming agents such as ethylene diamine tetraacetic acid, tartrate and citrate buffers, and high-molecular weight polymers such as polyethylene oxide for viscosity regulation may be added. Such compositions may be injected intramuscularly, intraperitoneally, or intravenously.
Further non-limiting examples of carriers and diluents include albumin and/or other plasma protein components such as low density lipoproteins, high density lipoproteins and the lipids with which these serum proteins are associated. These lipids include phosphatidyl choline, phosphatidyl serine, phosphatidyl ethanolamine and neutral lipids such as triglycerides. Lipid carriers also include, without limitation, tocopherol.
At least one polyalkylene oxide conjugated sFv linked to a therapeutic agent according to the invention may be administered by any means that achieve their intended puφose, for example, to treat various pathologies, such as cell inflammatory, allergy, tissue damage or other related pathologies.
A typical regimen for preventing, suppressing, or treating various pathologies comprises administration of an effective amount of an sFv conjugate, administered over a period of one or several days, up to and including between one week and about 24 months.
It is understood that the dosage ofthe present invention administered in vivo or in vitro will be dependent upon the age, sex, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature ofthe effect desired. The ranges of effective doses provided below are not intended to limit the invention and represent preferred dose ranges. However, the most preferred dosage will be tailored to the individual subject, as is understood and determinable by one of skill in the art, without undue experimentation. See, e.g., Berkow et al, eds., Merck Manual, 16th edition, Merck and Co., Rahway, NJ. (1992); Goodman etal., eds., Goodman andGilman's The Pharmacological Basis of Therapeutics, 8th edition, Pergamon Press, Inc., Elmsford, N. Y. ( 1990); Avery's Drug Treatment: Principles and Practice of Clinical Pharmacology and Therapeutics, 3rd edition, ADIS Press, LTD., Williams and Wilkins, Baltimore, MD. (1987), Ebadi, Pharmacology, Little, Brown and Co., Boston (1985), Katzung, Basic and Clinical Phamacology, Appleton and Lange, Norwalk, Conn. (1992), which references and references cited therein, are entirely incoφorated herein by reference.
The total dose required for each treatment may be administered by multiple doses or in a single dose. Effective amounts of a diagnostic/pharmaceutical compound or composition of the present invention are from about 0.001 μg to about 100 mg/kg body weight, administered at intervals of 4-72 hours, for a period of 2 hours to 5 years, or any range or value therein, such as 0.01-1.0, 1.0- 10, 10-50 and 50-100 mg/kg, at intervals of 1-4, 6-12, 12-24 and 24-72 hours, for a period of 0.5, 1.0-2.0, 2.0-4.0 and 4.0-7.0 days, or 1, 1-2, 2-4, 4-52 or more weeks, or 1, 2, 3-10, 10-20, 20-60 or more years, or any range or value therein. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions, which may contain auxiliary agents or excipients which are known in the art. Pharmaceutical compositions such as tablets and capsules can also be prepared according to routine methods. See, e.g. , Berker, supra, Goodman, supra, Avery, supra and Ebadi, supra, which are entirely incoφorated herein by reference, including all references cited therein.
Pharmaceutical compositions comprising at least one type of sFv conjugate of the invention, or, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 types of sFv conjugates, ofthe present invention may be contained in an amount effective to achieve its intended puφose. In addition to at least one sFv conjugate, a pharmaceutical composition may contain suitable pharmaceutically acceptable carriers, such as excipients, carriers and/or auxiliaries which facilitate processing ofthe active compounds into preparations which can be used pharmaceutically.
Pharmaceutical compositions may also include suitable solutions for administration intravenously, subcutaneously, dermally, orally, mucosally or rectally, and contain from about 0.01 to 99 percent, preferably from about 20 to 75 percent of active component (i.e., the sFv) together with the excipient. Pharmaceutical compositions for oral administration include tablets and capsules. Compositions which can be administered rectally include suppositories. See, e.g., Berker, supra, Goodman, supra, Avery, supra and Ebadi, supra. Additional lipid and lipoprotein drug delivery systems that may be included herein are described more fully in Annals N Y. Acad. Sci. 507:775-88, 98-103, and 252-271, which disclosure is hereby incoφorated by reference.
The compositions may also be formulated into orally administrable compositions containing one or more physiologically compatible carriers or excipients, and may be solid or liquid in form. These compositions may, if desired, contain conventional ingredients such as binding agents, for example, syrups, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, such as lactose, mannitol, starch, calcium phosphate, sorbitol, cyclodextran, or methylcellulose; lubricants such as magnesium stearate, high molecular weight polymers such as polyethylene glycols, high molecular weight fatty acids such as stearic acid or silica; disintegrants such as starch; acceptable wetting agents as, for example, sodium lauryl sulfate.
The oral compositions may assume any convenient form, such as tablets, capsules, lozenges, aqueous or oily suspensions, emulsions, or dry products suitable for reconstitution with water or other liquid medium prior to use. The liquid oral forms may, of course, contain flavors, sweeteners, preservatives such as methyl or propyl p-hydroxybenzoates; suspending agents such as sorbitol, glucose or other sugar syrup, methyl, hydroxymethyl, or carboxymethyl celluloses or gelatin; emulsifying agents such as lecithin or sorbitan monooleate or thickening agents. Non-aqueous compositions may also be formulated which comprise edible oils as, for example, fish-liver or vegetable oils. These liquid compositions may conveniently be encapsulated in, for example, gelatin capsules in a unit dosage amount.
The pharmaceutical compositions according to the present invention may also be administered, if appropriate, either topically as an aerosol or, formulated with conventional bases as a cream or ointment.
The pharmaceutical compositions of the present invention can also be administered by incoφorating the active ingredient into colloidal carriers, such as liposomes. Liposome technology is well known in the art, having been described by Allison et al, Nature 252:252-254 (1974), and Dancy et al, J. Immunol. 720:1109-1113 (1978).
Having now generally described this invention, the same will be better understood by reference to certain specific examples, which are included for the puφose of illustration and not intended to be limiting unless otherwise specified. Examples
Example 1 Preparation of Methoxypoly(ethylene glycolj-succinimidyl carbonate (SC-PEG)
Dissolve 60 g of methoxy poly(ethylene glycol) (MW 5,000) in 200 ml of 3/1 toluene/dichloromethane and treat with a toluene solution of phosgene (30 ml, 57 mmol) overnight. Evaporate the solution to dryness and remove the remainder of the phosgene under vacuum. Redissolve the residue in 150 ml of 2/1 toluene/dichloromethane. Treat the resulting solution with 2J g (18 mmol) of solid N-hydroxysuccinimide, followed by 1.7 ml (12 mmol) of triethylamine. Allow the solution to stand for three hours .and then filter it and evaporate it to dryness. Dissolve the residue in 600 ml of warm (50° C) ethyl acetate, filter the solution, and cool it to facilitate precipitation ofthe polymer. Collect the product by filtration, then recrystallize from ethyl acetate, and dry under vacuum over P2O5.
Example 2 Preparation ofCC 49/212 SCA
In the production of monovalent or multivalent antigen-binding proteins, the same recombinant E. coli production system that was used for prior single- chain antigen-binding protein production was used. See Bird et al, Science 242:423 (1988). This production system produced between 2 and 20% of the total E. coli protein as single-chain antigen-binding protein. For protein recovery , the frozen cell paste from three 10-liter fermentations (600-900 g) was thawed overnight at 4°C and gently resuspended at 4°C in 50 mM Tris-HCl, 1.0 mM EDTA, 100mM KCl, 0J mMPMSF,pH 8.0 (lysis buffer), using 10 liters of lysis buffer for every kilogram of wet cell paste. When thoroughly resuspended, the chilled mixture was passed three times through a Manton-Gaulin cell homogenizer to totally lyse the cells. Because the cell homogenizer raised the temperature of the cell lysate to 25 ± 5°C, the cell lysate was cooled to 5 + 2°C with a Lauda/Brinkman chilling coil after each pass. Complete lysis was verified by visual inspection under a microscope. The cell lysate was centrifuged at 24,300g for 30 minutes at 6°C using a
Sorvall RC-5B centrifuge. The pellet containing the insoluble single-chain antigen-binding protein was retained, and the supernatant was discarded. The pellet was washed by gently scraping it from the centrifuge bottles and resuspending it in 5 liters of lysis buffer/kg of wet cell paste. The resulting 3.0- to 4.5-liter suspension was again centrifuged at 24,300g for 30 minutes at 6°C, and the supernatant was discarded. This washing ofthe pellet removes soluble E. coli proteins and can be repeated as many as five times. At any time during this washing procedure the material can be stored as a frozen pellet at -20°C. A substantial time saving in the washing steps can be accomplished by utilizing a Pellicon tangential flow apparatus equipped with 0.22-μm microporous filters, in place of centrifugation.
The washed pellet was solubilized at 4°C in freshly prepared 6 M guanidine hydrochloride, 50 mM Tris-HCl, 10 mM CaCl2, 50 mM KC1, pH 8.0 (dissociating buffer), using 9 ml/g of pellet. If necessary, a few quick pulses from a Heat Systems Ultrasonics tissue homogenizer can be used to complete the solubilization. The resulting suspension was centrifuged at 24,300g for 45 minutes at 6°C and the pellet was discarded. The optical density of the supernatant was determined at 280 nm and if the OD2g0 was above 30, additional dissociating buffer was added to obtain an OD280 of approximately 25. The supernatant was slowly diluted into cold (4-7°C) refolding buffer (50 mM Tris-HCl, 10 mM CaCl2, 50 mM KC1, pH 8.0) until a 1 :10 dilution was reached (final volume 10 - 20 liters). Re-folding occurs over approximately eighteen hours under these conditions. The best results are obtained when the GuHCl extract is slowly added to the refolding buffer over a two hour period, with gentle mixing. The solution was filtered through a 0.2 μm Millipore Millipak 200. This filtration step may be optionally preceded by a centrifugation step. The filtrate was concentrated to 1 to 2 liters using an .Amicon spiral cartridge with 10,000 MWCO cartridge, again at 4°C.
The concentrated crude antigen-binding protein sample was dialyzed against Buffer G (60 mM MOPS, 0.5 mM Ca acetate, pH 6.0 - 6.4) until the conductivity was lowered to that of Buffer G. The sample was then loaded on a 21.5 x 250-mm polyaspartic acid PolyCAT A column, manufactured by Poly LC of Columbia, Maryland. If more than 60 mg of protein is loaded on this column, the resolution begins to deteriorate; thus, the concentrated crude sample often must be divided into several PolyCAT A runs. Most antigen-binding proteins have an extinction coefficient of about 2.0 ml mg 'cm"1 at 280 nm and this can be used to determine protein concentration. The antigen-binding protein sample was eluted from the PolyCAT A column with a 50-min linear gradient from Buffer G to Buffer H (60 mM MOPS, 20 mM Ca Acetate, pH 7.5 - 8.0). Most of the single-chain proteins elute between 20 and 26 minutes when this gradient is used. This corresponds to an eluting solvent composition of approximately 70% Buffer G and 30% Buffer H. Most ofthe bivalent antigen-binding proteins elute later than 45 minutes, which correspond to over 90%) Buffer H.
Example 3 Modification ofCC 49/212 Single Chain Antigen Binding Molecule
With SC-PEG
A sample of CC 49/212 single chain antigen binding molecule (MW=27000) dissolved in KPO4/NaCl buffer (pH 7.2) was obtained as described in Example 2. The protein was found to be pure using SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) and size exclusion chromatography. The concentration of the protein was 0.79 mg/ml. It was further concentrated to at least 2 mg/ml using an Amicon concentrator with a 10,000 dalton nominal size cut-off, i.e., anything greater than 10K is retained. The modification reaction, i.e., the coupling of the SC-PEG to the CC 49/212, was carried out in 50 mM KPO4, 150 mM NaCl buffer, which was the storage buffer the protein was supplied in. The pH was raised from 7.2 to 7.5. SC-PEG (MW 5,000) was added in a 50x molar excess to protein. At specific time intervals, the coupling reaction was terminated by the addition of a 5 Ox molar excess of glycine and the extent and progress of the coupling reaction was checked as a function of time using both size exclusion chromatography using a DuPont Zorbax 250 column and SDS-PAGE.
Free SC-PEG remaining in the samples was removed by extensive dialysis on an .Amicon Centricon 10.
The samples were checked for degree of modification by size exclusion chromatography. After concentrating the samples, the residual amine concentration on the protein was determined by titration with trinitrobenzene sulfonate and the percentage of amine groups that had reacted with the SC-PEG (the "% modification") was calculated from the results.
Dansyl derivatives of native single chain antigen binding molecule (CC 49/212) and hemoglobin, PEG SCA and hemoglobin, and N-acetyl lysine were prepared. These samples were then analyzed for amino acid. The results of this experiment are shown in Table 3.
Figure imgf000066_0001
Example 4 Competition ELISA
The CC49 monoclonal antibody was developed by Dr. Jeffrey Schlom's group, Laboratory of Tumor Immunology and Biology, National Cancer Institute. It binds specifically to the pan-carcinoma tumor antigen TAG-72. See Muraro, R. et al, Cancer Research 48: 4588-4596 (1988).
Figure 1 is a graphical representation of three competition ELISA's in which unlabeled PEG modified CC49/212 single-chain Fv (closed squares), CC49/212 single-chain Fv (open squares), CC49 IgG (open circles), and MOPC- 21 IgG (+) competed against a CC49 IgG radiolabeled with 125I for binding to the TAG-72 antigen on a human breast melanoma extract. MOPC-21 is a control antibody that does not bind to TAG-72 antigen. In this experiment, 50% competition of 125I-CC49 IgG binding required about 200 nM of CC49 IgG, about 550 nM of CC49/212 sFv, and about 3000 nM of PEG modified CC49/212 sFv. Example 5 Preparation of U-PEG-OH
Figure imgf000067_0001
Materials
Methoxypolyethylene glycol) (m-PEG) was obtained from Union Carbide. The solvents were obtained from Aldrich Chemical of Milwaukee, Wisconsin. The methoxypoly(ethylene glycol)-N-succinimidyl carbonate (SC-PEG) was prepared as described in U.S. Patent No. 5,122,614, using m-PEG having a molecular weight of about 5,000. Each of the products prepared in Examples 5-10 was confirmed structurally by carbon- 13 NMR. The branched polymer, U-PEG-OH, was prepared by adding 100 mg ( 1 J mmol) of 1 ,3-diamino-2-propanol to a solution of 10.0 g (2 mmol) of SC-PEG in 50 mL of methylene chloride. The mixture was stirced for 18 hours at room temperature then filtered. Excess solvent was removed by distillation in vacuo. The residue was recrystallized from 2-propanol to yield 7J g of product (70% yield).
Example 6 Preparation of U-PNP-PEG
Figure imgf000067_0002
The compound of Example 5 was activated with p-nitrophenyl chloroformate. First, 5.0g (0.5 mmol) of U-PEG-OH was azeotropically dried by refiuxing in 75 mL of toluene for 2 hours, resulting in the removal of 25 mL of solvent/water. The reaction mixture was cooled to 30 ° C, followed by the addition of 120 mg (0.6 mmol) of p-nitrophenyl chloroformate and 50 mg (0.6 mmol) of pyridine. The resulting mixture was stined for two hours at 45 °C, followed by stirring overnight at room temperature.
The reaction mixture was then filtered through CELITE™ , followed by removal ofthe solvent from the filtrate by distillation in vacuo. The residue was recrystallized from 2-propanol to yield 4.2 g (81% yield) ofthe product.
Example 7 Preparation of US-PEG
Figure imgf000068_0001
In this example, the U-PNP-PEG of Example 6 was reacted with N-hydroxysuccinimide to form the succinimidyl carbonate ester of U-PEG. A solution containing 5.0 g (0.5 mmol) of the U-PNP-PEG, 0.6 g (5 mmol) of N-hydroxysuccinimide and 0J3 g (1 mmol) of diisopropylethylamine in 40 ml of methylene chloride was refluxed for 18 hours. The solvent was then removed by distillation in vacuo, and the residue was recrystallized from 2-propanol to yield 4.2 g ofthe succinimidyl carbonate ester (82% yield). Example 8 Preparation of XU-PEG-OH
Figure imgf000069_0001
This branched polymer was prepared by reacting the U-PNP-PEG of Example 6 with 2-(2-aminoethoxy) ethanol (i.e., the amino alcohol was reacted with the p-nitrophenyl carbonate). The recrystallized product yield was 86%.
Example 9 Preparation of XU-PNP-PEG
The compound of Example 8 was functionalized with p-nitrophenyl carbonate as in Example 6. The recrystallized product yield was 83%.
Example 10 Preparation of XUS-PEG
Figure imgf000069_0002
In this example, the succinimidyl carbonate derivative of compound prepared in Example 8 was prepared according to the process described in Example 7, by reacting N-hydroxysuccinimide with the p-nitrophenyl carbonate derivative of Example 9. The recovered product yield was 84%.
Example 11 Modification ofCC49/218 with SC-PEG or XUS-PEG
A sample containing CC49/218 was desalted on a PD-10 column in a buffer consisting of OJM sodium phosphate, pH 8.0. .An equimolar amount of
SC-PEG or XUS-PEG was added and the reactions were incubated at 4°C, overnight. The reactions were quenched with an excess of glycine. The modified CC49/218 conj ugates were GPC purified and then concentrated in a centricon- 10.
The yield based on GPC integration was about 50% for SC-PEG modified
CC49/218 and about 40% for XUS-PEG modified CC49/218. The GPC profiles were almost identical to those obtained when the reaction was performed at pH9.0, room temperature. SDS-PAGE revealed that the appropriate derivatives had been made.
Example 12 Competition ELISA
The assay was performed as in Example 4 above using SC-PEG modified CC49/218 and XUS-PEG modified CC49/218 along with the appropriate controls. The results are shown in Figure 4 and in Table 4 below.
Figure imgf000071_0001
Thus, the affinity ofthe SC-PEG modified CC49-SCA was within about 8 to 10 fold of the native CC49-SCA and the affinity the XUS-PEG modified CC49-SCA was within about 4 to 5 fold ofthe native CC49-SCA.
Samples #049304 and #049303 were PEG modified, whiles samples #04901 and #049302 were unmodified CC49/218 isolated from the reaction mixtures.
Example 13 Pharmacokinetics of Plasma Retention of Fv and PEG-sFv
Sixty μg of CC49/218 sFv protein or 60 μg of PEG-modified sFv protein were injected intravenously at time 0 into ICR (CD- 1 ) female mice (Harlan - 25g, 7-8 weeks old). Mice were bled at the time points indicated in Figure 5. The percent retention in plasma was quantitated by ELISA methods. For the PEG- modified conjugate, CC49/218 sFv was conjugated to SC-PEG of molecular mass 20,000 (the protocol is described in U.S. Patent 5,122,614, which disclosure is incoφorated herein by reference). The average PEG:sFv molar ratio in the tested PEG-sFv conjugate was approximately 1 :1. Example 14 PEGylated Multimer Single Chain Antibodies
Using bifunctional PEG, dimers and trimers of CC49-SCA have been made. CC49-SCA was modified with bifunctional PEG as follows: 2 ml of CC49- SCA in phosphate buffered saline (25 mM sodium phosphate, pH 7.3, 0J5 M NaCl) at a concentration of 1.5 mg/ml was modified as follows. Bifunctional PEG (polyethylene glycol with reactive SC at both terminal ends), 1.887 mg (powder) was dissolved in OJ ml of MOPS (3-[N-moφholino]propane)-sulfonic acid) buffered at pH 7.3. This PEG solution was added to CC49-SCA solution within 10 seconds of dissolution. The mixture was then stirred at 24° C for 1 hour. At the end ofthe reaction, solid Guanidine HCI was added to the reaction mixture to a final concentration of 6 M in order to break up non covalently associated CC49- SC A. This material was immediately applied to a size exclusion column (2 cm X 60 cm, Superdex-75) previously equilibrated in the buffer composed of 60 mM Guanidine HCI in 50 mM Tris pH 7.3, ImM CaCl2, 0.1 mM PMSF (phenyl methyl sulfonyl flouride), and 50 mM KC1. Multimers of different molecular weights were then fractionated from the column.
These multimers are CC49-SCA separated by a long stretch of PEG (5000 MW, about 226 carbons in length) and were freshly refolded and thus, are not believed to arise from aggregation. It is less likely that there was diabody or multivalent SCA formed as a result of self association ofthe CC49-SCA. Further evidence was demonstrated by the fact that there was negligible native CC49-SC A in the denaturing SDS-PAGE profile.
The SDS-PAGE electrophoresis patterns for dimeric CC49-SCA, trimeric CC49-SCA, PEG-CC49-SCA and native CC49-SCA under reducing conditions are shown in Figure 6.
The multimers were assayed for binding affinity using the following assay which was modified method described in B Friquet et al. J. of Immunology Methods, 77:305-319 ( 1985). Briefly, various amounts of a given modified single chain antibody were mixed with various amounts ofthe antigen mucin in PBS (phosphate buffered saline). The binding reaction was allowed to reach equilibration for at least 24 hours at 4°C. At the end of the incubation, the unbound CC49-SCA fractions were assayed by ELISA, while the bound fractions were washed away. The total amount of free CC49-SC A was determined by the ELISA using the CC49-SCA sample pre-incubated in the absence ofthe antigen mucin. The bound antibody was determined by subtraction ofthe free (unbound) amount from the total amount as determined by ELISA. Since the total amount ofthe CC49-SCA was known, it was also used as its own standard curve. Note that each type of CC49-SCA had its own reference control. In essence, the protocol was measuring the unbound amount of a particular version of PEG- CC49-SC A as a result ofthe binding to the antigen. Although PEG may affect the detection reagent in ELISA, this was well contained in the standard references. Therefore, the amount measured was not due to the difference of various PEG on the various versions of PEG-CC49-SCA.
In sum, in this study, increasing concentrations ofthe CC49-SCA protein were allowed to bind to a fixed amount ofthe antigen. The amount that binds to 50% ofthe maximal level is a good indication ofthe affinity. This data indicate that the affinity of PEG-Di-CC49-SCA and PEG-Tri-CC49-SCA are very similar to that of native CC49-SCA. However, the PEG-modified CC49-SCA monomer had much lower affinity. The binding data are shown in Figure 7.
Example 15 Pharmacokinetics ofPEG-CC49-SCA
The pharmacokinetic study of various forms of PEG-CC49-SCA was performed as in Example 13, above.
The data obtained from this study indicate the following: There was a trend toward longer circulation half-lives as the size ofthe
PEG increases. As more PEG is attached to the protein, the circulation time increases. However, attachment of a few strands of high molecular weight PEG gives a better increase in circulation half-life than multiple stands of lower molecular weight PEG.
The circulation half-life of CC49-SCA-U-PEG, made with the US-PEG prepared in Example 7, was about the same as that of CC49-SCA-PEG- 12000. Therefore, the shape ofthe PEG does not affect the circulation half-life.
Whether the linker is an SC-bond, Flan-bond, hydrazine bond, or TPC, there was no significant change in the circulation half-life. Therefore, the chemical bonds of the linkers, if not releasable, do not affect the circulation half-life. In addition, the PEG remains attached to the protein during the observable time.
The circulation half-life was shortened by carbohydrate. However, if PEG was attached to the carbohydrate, it increases the circulation by about 10 fold. This was not better, however, than attaching an equivalent number of PEG at other sites on CC49-SCA.
The results ofthe study are shown in the table below:
Pharmacokinetic Data of SCA and PEG-SCA
Figure imgf000075_0001
NOTES:
A. The PK modeling was following a one-compartment, i.v.-bolus model.
B. The observed vs. predicted correlations are over 95%.
C. The circulatory half-life is the time for the drug concentration in the serum to reduce to one half after equilibration is reached (about 2 min.)
D. AUC = area under the curve, is the integral ofthe drug blood level over time from zero to last measurement, and is a measure of quantity of drug absorbed and in the body.
E. Mean residence time is the average amount of time a drug remains in the compartment.
F. The code for linkers are:
SC = Succinimidyl Carbonate NHS = N-hydroxy succinimide
Hz = Hydrazide TPC = trichlorophenyl carbonate
Flan = Thiazolidine thione ester U = SC in the middle of PEG
The Flan-bond PEG was prepared as taught in U.S. Patent 5,405,877.
Example 16 Competition Binding Assay
A competition binding assay of biotinylated CC49-SCA with various PEGylated CC49-SCA proteins was performed using an ELISA ofthe biotinylated CC49-SCA as detected by horseradish-peroxidase conjugated with streptoavidin
(SAN-HRP). In the assay, the biotinylated CC49-SCA and PEG-CC49-SCA sample were mixed at various ratios for competition of binding to the antigen mucin on a surface. The amount of biotinylated-CC49-SCA bound to antigen was then measured by SAN-HRP, which would not detect the PEG-modified CC49- SCA. The reduction of binding of biotinylated CC49-SCA due to competition of
PEG-CC49-SCA is a reflection ofthe relative affinity ofthe two forms of CC49- SCA for the antigen. The level of PEG-CC49-SCA that caused reduction of biotin-CC49-SCA to half of its maximum binding level (IC50) was used in the Cheng-Prussoff formula for determination ofthe affinity constant Kd as follows: Assuming the biotin-CC49-SCA level is [s] and its affinity is known as Ks, then the affinity for the PEG-CC49-SCA is Kd = IC50/(1 + [s]/Ks). Note that PEG- CC49-SCA was not directly measured and that PEG had no effect on biotin binding because it was on a different molecule. The estimated Kd was then expressed as a percentage ofthe control (CC49-SCA). The affinity ranking obtained was as follows:
CC49 = SC2 > GC = Bio.CC49 = CC12 > C20 = F5 >HZ >PG where SC2 is PEG SC2000-CC49-SCA; GC is glyco-CC49-SCA; Bio.CC49-SCA is the biotinylated CC49-SCA; CC12 is the PEG-SC12,000-CC49-SCA; F5 is PEG-Flan-5000-CC49-SCA; HZ is CC49-SCA highly PEGylated on the carboxyl groups with MW 5000 hydrazine-PEG; PG is PEG-glyco-CC49-SCA and is highly PEGylated on the carbohydrate with MW 5000 hydrazine PEG; and C20 is PEG-SC20000-CC49-SC A. The affinity for all the PEG-modified CC49-SCAs shown in Figure 8, were within about two fold ofthe native CC49-SCA. Taken together with the data in Example 15 (see, the table), the results indicate that when the SCA is modified with a lower number of PEG molecules, the resulting affinity ofthe SCA is better than when the SCA is modified with a higher number of PEG molecules.
Although the foregoing refers to particular prefened embodiments, it will be understood that the present invention is not so limited. It will occur to those skilled in the art that various modifications may be made to the disclosed embodiments and that such modifications are intended to be within the scope of the present invention.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: ENZON, INC.
20 KINGSBRIDGE ROAD PISCATAWAY, NJ 08854-3963 UNITED STATES OF AMERICA
APPLICANTS/INVENTORS: WHITLOW, MARC
SHORR, ROBERT G.L. FILPULA, DAVID R. LEE, LIHSYNG S.
(ii) TITLE OF INVENTION: POLYALKYLENE OXIDE-MODIFIED SINGLE CHAIN POLYPEPTIDES
(iii) NUMBER OF SEQUENCES: 4
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: STERNE, KESSLER, GOLDSTEIN & FOX P.L.L.C.
(B) STREET: 1100 NEW YORK AVENUE, SUITE 600
(C) CITY: WASHINGTON
(D) STATE: DC
(E) COUNTRY: USA
(F) ZIP: 20005
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.30
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: To be assigned
(B) FILING DATE: Herewith
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 60/044,449
(B) FILING DATE: 30-APR-1997
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 60/050,472
(B) FILING DATE: 23-JUN-1997
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 60/063,074
(B) FILING DATE: 27-OCT-1997
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 60/067,341
(B) FILING DATE: 02-DEC-1997
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: JORGE A. GOLDSTEIN
(B) REGISTRATION NUMBER: 29,021
(C) REFERENCE/DOCKET NUMBER: 0977.184PC02
(ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: 202-371-2600 (B) TELEFAX: 202-371-2540
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 749 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: both
(ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..738
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
GAC GTC GTG ATG TCA CAG TCT CCA TCC TCC CTA CCT GTG TCA GTT GGC 48
Asp Val Val Met Ser Gin Ser Pro Ser Ser Leu Pro Val Ser Val Gly
1 5 10 15
GAG AAG GTT ACT TTG AGC TGC AAG TCC AGT CAG AGC CTT TTA TAT AGT 96
Glu Lys Val Thr Leu Ser Cys Lys Ser Ser Gin Ser Leu Leu Tyr Ser
20 25 30
GGT AAT CAA AAG AAC TAC TTG GCC TGG TAC CAG CAG AAA CCA GGG CAG 144
Gly Asn Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys Pro Gly Gin
35 40 45
TCT CCT AAA CTG CTG ATT TAC TGG GCA TCC GCT AGG GAA TCT GGG GTC 192
Ser Pro Lys Leu Leu lie Tyr Trp Ala Ser Ala Arg Glu Ser Gly Val 50 55 60
CCT GAT CGC TTC ACA GGC AGT GGA TCT GGG ACA GAT TTC ACT CTC TCC 240
Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Ser
65 70 75 80
ATC AGC TGT GTG AAG ACT GAA GAC CTG GCA GTT TAT TAC TGT CAG CAG 288 lie Ser Cys Val Lys Thr Glu Asp Leu Ala Val Tyr Tyr Cys Gin Gin
85 90 95
TAT TAT AGC TAT CCC CTC ACG TTC GGT GCT GGG ACC AAG CTT GTG CTG 336
Tyr Tyr Ser Tyr Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Val Leu
100 105 110
AAA GGC TCT TGT TCC GGT AGC GGC AAA CCC GGG AGT GGT GAA GGT AGC 384
Lys Gly Ser Cys Ser Gly Ser Gly Lys Pro Gly Ser Gly Glu Gly Ser
115 120 125
ACT AAA GGT CAG GTT CAG CTG CAG CAG TCT GAC GCT GAG TTG GTG AAA 432
Thr Lys Gly Gin Val Gin Leu Gin Gin Ser Asp Ala Glu Leu Val Lys 130 135 140
CCT GGG GCT TCA GTG AAG ATT TCC TGC AAG GCT TCT GGC TAC ACC TTC 480 Pro Gly Ala Ser Val Lys lie Ser Cys Lys Ala Ser Gly Tyr Thr Phe
145 150 155 160
ACT GAC CAT GCA ATT CAC TGG GTG AAA CAG AAC CCT GAA CAG GGC CTG 528 Thr Asp His Ala lie His Trp Val Lys Gin Asn Pro Glu Gin Gly Leu 165 170 175
GAA TGG ATT GGA TAT TTT TCT CCC GGA AAT GAT GAT TTT AAA TAC AAT 576
Glu Trp lie Gly Tyr Phe Ser Pro Gly Asn Asp Asp Phe Lys Tyr Asn 180 185 190
GAG AGG TTC AAG GGC AAG GCC ACA CTG ACT GCA GAC AAA TCC TCC AGC 624
Glu Arg Phe Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser 195 200 205
ACT GCC TAC GTG CAG CTC AAC TGC CTG ACA TCT GAG GAT TCT GCA GTG 672
Thr Ala Tyr Val Gin Leu Asn Cys Leu Thr Ser Glu Asp Ser Ala Val 210 215 220
TAT TTC TGT ACA AGA TCC CTG AAT ATG GCC TAC TGG GGT CAA GGA ACC 720
Tyr Phe Cys Thr Arg Ser Leu Asn Met Ala Tyr Trp Gly Gin Gly Thr
225 230 235 240
TCA GTC ACC GTC TCC TGC TAATAGGATC C 749
Ser Val Thr Val Ser Cys 245
(2) INFORMATION FOR SEQ ID NO : 2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 246 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 2 :
Asp Val Val Met Ser Gin Ser Pro Ser Ser Leu Pro Val Ser Val Gly 1 5 10 15
Glu Lys Val Thr Leu Ser Cys Lys Ser Ser Gin Ser Leu Leu Tyr Ser 20 25 30
Gly Asn Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys Pro Gly Gin 35 40 45
Ser Pro Lys Leu Leu lie Tyr Trp Ala Ser Ala Arg Glu Ser Gly Val 50 55 60
Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Ser 65 70 75 80
He Ser Cys Val Lys Thr Glu Asp Leu Ala Val Tyr Tyr Cys Gin Gin 85 90 95
Tyr Tyr Ser Tyr Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Val Leu 100 105 110
Lys Gly Ser Cys Ser Gly Ser Gly Lys Pro Gly Ser Gly Glu Gly Ser 115 120 125
Thr Lys Gly Gin Val Gin Leu Gin Gin Ser Asp Ala Glu Leu Val Lys 130 135 140 Pro Gly Ala Ser Val Lys He Ser Cys Lys Ala Ser Gly Tyr Thr Phe 145 150 155 160
Thr Asp His Ala He His Trp Val Lys Gin Asn Pro Glu Gin Gly Leu 165 170 175
Glu Trp He Gly Tyr Phe Ser Pro Gly Asn Asp Asp Phe Lys Tyr Asn 180 185 190
Glu Arg Phe Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser 195 200 205
Thr Ala Tyr Val Gin Leu Asn Cys Leu Thr Ser Glu Asp Ser Ala Val 210 215 220
Tyr Phe Cys Thr Arg Ser Leu Asn Met Ala Tyr Trp Gly Gin Gly Thr 225 230 235 240
Ser Val Thr Val Ser Cys 245
(2) INFORMATION FOR SEQ ID NO : 3 :
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 782 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: both
(D) TOPOLOGY: both
(ll) MOLECULE TYPE: cDNA
FEATURE :
(A) NAME/KEY: CDS
(B) LOCATION: 1..771
(xi ) SEQUENCE DESCRIPTION: SEQ ID NO : 3 :
GAC GTC GTG ATG TCA CAG TCT CCA TCC TCC CTA CCT GTG TCA GTT GGC 48
Asp Val Val Met Ser Gin Ser Pro Ser Ser Leu Pro Val Ser Val Gly 250 255 260
GAG AAG GTT ACT TTG AGC TGC AAG TCC AGT CAG AGC CTT TTA TAT AGT 96
Glu Lys Val Thr Leu Ser Cys Lys Ser Ser Gin Ser Leu Leu Tyr Ser 265 270 275
GGT AAT CAA AAG AAC TAC TTG GCC TGG TAC CAG CAG AAA CCA GGG CAG 144
Gly Asn Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys Pro Gly Gin
280 285 290
TCT CCT AAA CTG CTG ATT TAC TGG GCA TCC GCT AGG GAA TCT GGG GTC 192
Ser Pro Lys Leu Leu He Tyr Trp Ala Ser Ala Arg Glu Ser Gly Val 295 300 305 310
CCT GAT CGC TTC ACA GGC AGT GGA TCT GGG ACA GAT TTC ACT CTC TCC 240
Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Ser 315 320 325
ATC AGC AGT GTG AAG ACT GAA GAC CTG GCA GTT TAT TAC TGT CAG CAG 288
He Ser Ser Val Lys Thr Glu Asp Leu Ala Val Tyr Tyr Cys Gin Gin 330 335 340 TAT TAT AGC TAT CCC CTC ACG TTC GGT GCT GGG ACC AAG CTT GTG CTG 336
Tyr Tyr Ser Tyr Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Val Leu 345 350 355
AAA GGC TCT ACT TCC GGT AGC GGC AAA CCC GGG AGT GGT GAA GGT AGC 384
Lys Gly Ser Thr Ser Gly Ser Gly Lys Pro Gly Ser Gly Glu Gly Ser 360 365 370
ACT AAA GGT CAG GTT CAG CTG CAG CAG TCT GAC GCT GAG TTG GTG AAA 432
Thr Lys Gly Gin Val Gin Leu Gin Gin Ser Asp Ala Glu Leu Val Lys
375 380 385 390
CCT GGG GCT TCA GTG AAG ATT TCC TGC AAG GCT TCT GGC TAC ACC TTC 480
Pro Gly Ala Ser Val Lys He Ser Cys Lys Ala Ser Gly Tyr Thr Phe 395 400 405
ACT GAC CAT GCA ATT CAC TGG GTG AAA CAG AAC CCT GAA CAG GGC CTG 528
Thr Asp His Ala He His Trp Val Lys Gin Asn Pro Glu Gin Gly Leu
410 415 420
GAA TGG ATT GGA TAT TTT TCT CCC GGA AAT GAT GAT TTT AAA TAC AAT 576
Glu Trp He Gly Tyr Phe Ser Pro Gly Asn Asp Asp Phe Lys Tyr Asn 425 430 435
GAG AGG TTC AAG GGC AAG GCC ACA CTG ACT GCA GAC AAA TCC TCC AGC 624
Glu Arg Phe Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser 440 445 450
ACT GCC TAC GTG CAG CTC AAC AGC CTG ACA TCT GAG GAT TCT GCA GTG 672
Thr Ala Tyr Val Gin Leu Asn Ser Leu Thr Ser Glu Asp Ser Ala Val
455 460 465 470
TAT TTC TGT ACA AGA TCC CTG AAT ATG GCC TAC TGG GGT CAA GGA ACC 720
Tyr Phe Cys Thr Arg Ser Leu Asn Met Ala Tyr Trp Gly Gin Gly Thr 475 480 485
TCG GTC ACC GTC TCC AAA AAG AAG AAA AAA AAG AAA AAG GTC ACC GTC 768
Ser Val Thr Val Ser Lys Lys Lys Lys Lys Lys Lys Lys Val Thr Val
490 495 500
TCC TAATAGGATC C 782
Ser
(2) INFORMATION FOR SEQ ID NO: 4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 257 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 4 :
Asp Val Val Met Ser Gin Ser Pro Ser Ser Leu Pro Val Ser Val Gly 1 5 10 15
Glu Lys Val Thr Leu Ser Cys Lys Ser Ser Gin Ser Leu Leu Tyr Ser 20 25 30 Gly Asn Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys Pro Gly Gin 35 40 45
Ser Pro Lys Leu Leu He Tyr Trp Ala Ser Ala Arg Glu Ser Gly Val 50 55 60
Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Ser 65 70 75 80
He Ser Ser Val Lys Thr Glu Asp Leu Ala Val Tyr Tyr Cys Gin Gin 85 90 95
Tyr Tyr Ser Tyr Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Val Leu 100 105 110
Lys Gly Ser Thr Ser Gly Ser Gly Lys Pro Gly Ser Gly Glu Gly Ser 115 120 125
Thr Lys Gly Gin Val Gin Leu Gin Gin Ser Asp Ala Glu Leu Val Lys 130 135 140
Pro Gly Ala Ser Val Lys He Ser Cys Lys Ala Ser Gly Tyr Thr Phe 145 150 155 160
Thr Asp His Ala He His Trp Val Lys Gin Asn Pro Glu Gin Gly Leu 165 170 175
Glu Trp He Gly Tyr Phe Ser Pro Gly Asn Asp Asp Phe Lys Tyr Asn 180 185 190
Glu Arg Phe Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser 195 200 205
Thr Ala Tyr Val Gin Leu Asn Ser Leu Thr Ser Glu Asp Ser Ala Val 210 215 220
Tyr Phe Cys Thr Arg Ser Leu Asn Met Ala Tyr Trp Gly Gin Gly Thr 225 230 235 240
Ser Val Thr Val Ser Lys Lys Lys Lys Lys Lys Lys Lys Val Thr Val 245 250 255
Ser

Claims

ClaimsWhat is claimed is:
1. A single-chain antigen-binding polypeptide-polyalkylene oxide conjugate, comprising a single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein said single-chain antigen-binding polypeptide is conjugated to polyalkylene oxide and wherein said single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within a range of about one-fold to about ten-fold of the antigen binding affinity of said single-chain antigen-binding polypeptide in its unconjugated form.
2. A single-chain antigen-binding polypeptide-polyalkylene oxide conjugate, comprising a single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein said single-chain antigen-binding polypeptide is conjugated to polyalkylene oxide and wherein said single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about ten-fold of the antigen binding affinity of said single-chain antigen-binding polypeptide in its unconjugated form.
3. A single-chain antigen-binding polypeptide-polyalkylene oxide conjugate, comprising a single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein said single-chain antigen-binding polypeptide is conjugated to polyalkylene oxide and wherein said single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about five-fold of the antigen binding affinity of said single-chain antigen-binding polypeptide in its unconjugated form.
4. A single-chain antigen-binding polypeptide-polyalkylene oxide conjugate, comprising a single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and (c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein said single-chain antigen-binding polypeptide is conjugated to polyalkylene oxide and wherein said single-chain antigen-binding polypeptide-polyalkylene oxide conjugate has an antigen binding affinity within about two-fold of the antigen binding affinity of said single-chain antigen-binding polypeptide in its unconjugated form.
5. A single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking said first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue which is capable of polyalkylene oxide conjugation, wherein said Cys residue is located at a position selected from the group consisting of:
(i) the amino acid position 11, 12, 13, 14 or 15 ofthe light chain variable region;
(ii) the amino acid position 77, 78 or 79 ofthe light chain variable region;
(iii) the amino acid position 11 , 12, 13 , 14 or 15 of the heavy chain variable region;
(iv) the amino acid position 82B, 82C or 83 of the heavy chain variable region;
(v) any amino acid position ofthe peptide linker;
(vi) adjacent to the C-terminus of polypeptide (a) or (b); and
(vii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
6. The single-chain antigen-binding polypeptide of claim 5 , wherein said Cys residue capable of polyalkylene oxide conjugation is located at a position selected from the group consisting of:
(i') the amino acid position 77 ofthe light chain variable region; (ii') the amino acid position 82B ofthe heavy chain variable region; (iii') the amino acid position 3 ofthe peptide linker; (iv') adjacent to the C-terminus of said polypeptide (a) or (b); and (v') combinations thereof.
7. The single-chain antigen-binding polypeptide of claim 5 , wherein said first polypeptide (a) comprises the antigen binding portion ofthe variable region of an antibody light chain and said second polypeptide (b) comprises the antigen binding portion ofthe variable region of an antibody heavy chain.
8. The single-chain antigen-binding polypeptide of claim 5 , wherein the C- terminus of said second polypeptide (b) is the native C-terminus.
9. The single-chain antigen-binding polypeptide of claim 5 , wherein the C- terminus of said second polypeptide (b) comprises a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues of the second polypeptide are sufficient for the polyalkylene oxide conjugated single-chain antigen-binding polypeptide to be capable of binding an antigen.
10. The single-chain antigen-binding polypeptide of claim 5 , wherein the C- terminus of said second polypeptide (b) comprises an addition of one or plurality of amino acid residue(s), such that the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
11. The single-chain antigen-binding polypeptide of claim 5, wherein said wherein said Cys residue capable of polyalkylene oxide conjugation is attached to a polyalkylene oxide moiety.
12. The polyalkylene oxide conjugated single-chain antigen-binding polypeptide of claim 11 , wherein said polyalkylene oxide conjugated single-chain antigen-binding polypeptide is conjugated to one or plurality of peptide, lipid, nucleic acid, drug, toxin, chelator, boron addend or detectable label molecules.
13. The polyalkylene oxide conjugated single-chain antigen-binding polypeptide of claim 11 , wherein said polyalkylene oxide conjugated single-chain antigen-binding polypeptide is conjugated to a carrier having one or plurality of peptide, lipid, nucleic acid, drug, toxin, chelator, boron addend or detectable label molecules bound to said carrier.
14. A polynucleotide sequence encoding a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking said first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue which is capable of polyalkylene oxide conjugation, wherein said Cys residue is located at a position selected from the group consisting of:
(i) the amino acid position 11, 12, 13, 14 or 15 of said light chain variable region; (ii) the amino acid position 77, 78 or 79 of said light chain variable region;
(iii) the amino acid position 11, 12, 13, 14 or 15 of said heavy chain variable region;
(iv) the amino acid position 82B, 82C or 83 of said heavy chain variable region;
(v) any amino acid position of said peptide linker;
(vi) adjacent to the C-terminus of said polypeptide (a) or (b); and
(vii) combinations thereof, and wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
15. The polynucleotide sequence of claim 14, wherein said Cys residue capable of polyalkylene oxide conjugation is located at a position selected from the group consisting of:
(i') the amino acid position 77 ofthe light chain variable region; (ii') the amino acid position 82B ofthe heavy chain variable region; (iii') the amino acid position 3 ofthe peptide linker; (iv') adjacent to the C-terminus of said polypeptide (a) or (b); and (v') combinations thereof.
16. A replicable cloning or expression vehicle comprising the polynucleotide sequence of claim 14.
17. The vehicle of claim 16 which is a plasmid.
18. A host cell transformed with the polynucleotide of claim 17.
19. The host cell of claim 18 which is a bacterial cell, a yeast cell or other fungal cell, an insect cell or a mammalian cell line.
20. The host cell of claim 19 which is Pichia pastoris.
21. A method of a producing single-chain antigen-binding polypeptide- polyalkylene oxide conjugate, comprising a single-chain antigen-binding polypeptide comprising:
(a) providing a first genetic sequence encoding a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) providing a second genetic sequence encoding a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) linking the first and second genetic sequences (a) and (b) with a third genetic sequence encoding a linker into a fourth genetic sequence encoding a single chain polypeptide having an antigen binding site, wherein said single-chain antigen-binding polypeptide is capable of polyalkylene oxide conjugation and wherein said single-chain antigen-binding polypeptide-polyalkylene oxide conjugate retains antigen binding affinity within a range of about one-fold to about ten-fold of the antigen binding affinity of said single-chain antigen-binding polypeptide in its unconjugated form.
22. A method of producing a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) providing a first genetic sequence encoding a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) providing a second genetic sequence encoding a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and (c) linking said first and second genetic sequences (a) and (b) with a third genetic sequence encoding a peptide linker into a fourth genetic sequence encoding a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least one Cys residue which is capable of polyalkylene oxide conjugation, wherein said Cys residue is located at a position selected from the group consisting of:
(i) the amino acid position 11, 12, 13, 14 or 15 of said light chain variable region;
(ii) the amino acid position 77, 78 or 79 of said light chain variable region;
(iii) the amino acid position 11, 12, 13, 14 or 15 of said heavy chain variable region;
(iv) the amino acid position 82B, 82C or 83 of said heavy chain variable region;
(v) any amino acid position of said peptide linker;
(vi) adjacent to the C-terminus of said polypeptide (a) or (b); and
(vii) combinations thereof, and wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen;
(d) transforming a host cell with said fourth genetic sequence encoding said single-chain antigen-binding polypeptide of step (c); and
(e) expressing said single-chain antigen-binding polypeptide of step (c) in said host, thereby producing a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation.
23. The method of claim 22, wherein said Cys residue capable of polyalkylene oxide conjugation is located at a position selected from the group consisting of:
(i') the amino acid position 77 ofthe light chain variable region;
(ii') the amino acid position 82B ofthe heavy chain variable region; (iii') the amino acid position 3 ofthe peptide linker;
(iv') adjacent to the C-terminus of said polypeptide (a) or (b); and
(v') combinations thereof.
24. The method of claim 22, said first genetic sequence encoding a first polypeptide (a) comprises the antigen binding portion ofthe variable region of an antibody light chain and said second genetic sequence encoding a second polypeptide (b) comprises the antigen binding portion ofthe variable region of an antibody heavy chain.
25. The method of claim 22, wherein the C-terminus of said second polypeptide (b) is the native C-terminus.
26. The method of claim 22, wherein the C-terminus of said second polypeptide (b) comprises a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe second polypeptide are sufficient for the polyalkylene oxide conjugated single-chain antigen-binding polypeptide to be capable of binding an antigen.
27. The method of claim 22, wherein the C-terminus of said second polypeptide (b) comprises an addition of one or plurality of amino acid residue(s), such that the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
28. A multivalent single-chain antigen-binding polypeptide-polyalkylene oxide conjugate, comprising two or more single-chain antigen binding polypeptides, each single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; (b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein said single-chain antigen-binding polypeptide is conjugated to polyalkylene oxide and wherein said single-chain antigen-binding polypeptide-polyalkylene oxide conjugate retains antigen binding affinity within a range of about one-fold to about ten-fold of the antigen binding affinity of said single-chain antigen-binding polypeptide in its unconjugated form.
29. A multivalent single-chain antigen-binding protein, comprising two or more single-chain antigen-binding polypeptides, each single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking said first and second polypeptides wherein one ofthe two single-chain antigen-binding polypeptide has at least one Cys residue which is capable of polyalkylene oxide conjugation, wherein said Cys residue is located at a position selected from the group consisting of:
(i) the amino acid position 11, 12, 13, 14 or 15 of said light chain variable region;
(ii) the amino acid position 77, 78 or 79 of said light chain variable region;
(iii) the amino acid position 11, 12, 13, 14 or 15 of said heavy chain variable region;
(iv) the amino acid position 82B, 82C or 83 of said heavy chain variable region; (v) any amino acid position of said peptide linker; (vi) adjacent to the C-terminus of said polypeptide (a) or (b); and (vii) combinations thereof, and wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
30. The multivalent protein of claim 29, wherein said Cys residue capable of polyalkylene oxide conjugation is located at a position selected from the group consisting of:
(i') the amino acid position 77 ofthe light chain variable region; (ii') the amino acid position 82B ofthe heavy chain variable region; (iii') the amino acid position 3 ofthe peptide linker; (iv') adjacent to the C-terminus of said polypeptide (a) or (b); and (v') combinations thereof.
31. The multivalent protein of claim 29, wherein said first polypeptide (a) comprises the antigen binding portion ofthe variable region of an antibody light chain and said second polypeptide (b) comprises the antigen binding portion ofthe variable region of an antibody heavy chain.
32. The multivalent protein of claim 29, wherein the C-terminus of said second polypeptide (b) is the native C-terminus.
33. The multivalent protein of claim 29, wherein the C-terminus of said second polypeptide (b) comprises a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe second polypeptide are sufficient for the polyalkylene oxide conjugated single-chain antigen-binding polypeptide to be capable of binding an antigen.
34. The multivalent protein of claim 29, wherein the C-terminus of said second polypeptide comprises an addition of one or plurality of amino acid residue(s), such that the polyalkylene oxide conjugated single-chain antigen- binding polypeptide is capable of binding an antigen.
35. The multivalent protein of claim 29, wherein said wherein said Cys residue capable of polyalkylene oxide conjugation is attached to a polyalkylene oxide moiety.
36. The polyalkylene oxide conjugated multivalent protein of claim 35, wherein said polyalkylene oxide conjugated multivalent protein is conjugated to one or plurality of peptide, lipid, nucleic acid, drug, toxin, chelator, boron addend or detectable label molecule(s).
37. A method of detecting an antigen suspected of being in a sample, comprising:
(a) contacting said sample with the polyalkylene oxide conjugated polypeptide or protein of claim 1 or 11, wherein said polyalkylene oxide conjugated polypeptide or protein is conjugated to one or plurality of detectable label molecule(s), or conjugated to a carrier having one or plurality of detectable label molecule(s) bound to said carrier; and
(b) detecting whether said polyalkylene oxide conjugated single-chain antigen-binding polypeptide or protein has bound to said antigen.
38. A method of imaging the internal structure of -an animal, comprising administering to said animal an effective amount of the polyalkylene oxide conjugated polypeptide or protein of claim 1 or 11, wherein said polyalkylene oxide conjugated polypeptide or protein is conjugated to one or plurality of detectable label or chelator molecule(s), or conjugated to a carrier having one or plurality of detectable label or chelator molecule(s) bound to said carrier, and measuring detectable radiation associated with said animal.
39. The method of claim 38, wherein said animal includes a human.
40. A method for treating a targeted disease, comprising administering an effective amount of a composition comprising the polyalkylene oxide conjugated polypeptide or protein of claim 1 or 11 and a pharmaceutically acceptable carrier vehicle, wherein said polyalkylene oxide conjugated polypeptide or protein is conjugated to one or plurality of peptide, lipid, nucleic acid, drug, toxin, boron addend or radioisotope molecule(s), or conjugated to a carrier having one or plurality of peptide, lipid, nucleic acid, drug, toxin, boron addend or radioisotope molecule(s) bound to said carrier.
41. A single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least three consecutive Lys residues which are capable of polyalkylene oxide conjugation and wherein any one of said consecutive Lys residues is located at a position selected from the group consisting of
(i) any amino acid position ofthe peptide linker;
(ii) adjacent to the C-terminus ofthe second polypeptide (b); and
(iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
42. A polynucleotide sequence encoding a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least three consecutive Lys residues which are capable of polyalkylene oxide conjugation and wherein any one of said consecutive Lys residues is located at a position selected from the group consisting of
(i) any amino acid position ofthe peptide linker;
(ii) adjacent to the C-terminus ofthe second polypeptide (b); and
(iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
43. A replicable cloning or expression vehicle comprising the polynucleotide sequence of claim 42.
44. The vehicle of claim 43 which is a plasmid.
45. A host cell transformed with the polynucleotide of claim 42.
46. The host cell of claim 39 which is a bacterial cell, a yeast cell or other fungal cell, an insect cell or a mammalian cell line.
47. The host cell of claim 46 which is Pichia pastoris.
48. A method of producing a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) a second polypeptide comprising the antigen binding portion of the variable region of an antibody heavy or light chain; and
(c) a peptide linker linking the first and second polypeptides (a) and (b) into a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least three consecutive Lys residues which are capable of polyalkylene oxide conjugation and wherein any one of said consecutive Lys residues is located at a position selected from the group consisting of
(i) any amino acid position ofthe peptide linker; (ii) adjacent to the C-terminus ofthe second polypeptide (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
49. The single-chain antigen-binding polypeptide of claim 41 , wherein said wherein said wherein any one of said consecutive Lys residues capable of polyalkylene oxide conjugation is attached to a polyalkylene oxide moiety.
50. The polyalkylene oxide conjugated single-chain antigen-binding polypeptide of claim 49, wherein said polyalkylene oxide conjugated single-chain antigen-binding polypeptide is conjugated to one or plurality of peptide, lipid, nucleic acid, drug, toxin, chelator, boron addend or detectable label molecules.
51. The polyalkylene oxide conjugated single-chain antigen-binding polypeptide of claim 49, wherein said polyalkylene oxide conjugated single-chain antigen-binding polypeptide is conjugated to a carrier having one or plurality of peptide, lipid, nucleic acid, drug, toxin, chelator, boron addend or detectable label molecules bound to said carrier.
52. A method of producing a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation, comprising:
(a) providing a first genetic sequence encoding a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain;
(b) providing a second genetic sequence encoding a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) linking said first and second genetic sequences (a) and (b) with a third genetic sequence encoding a peptide linker into a fourth genetic sequence encoding a single chain polypeptide having an antigen binding site, wherein the single-chain antigen-binding polypeptide has at least three consecutive Lys residues which are capable of polyalkylene oxide conjugation and wherein any one of said consecutive Lys residues is located at a position selected from the group consisting of
(i) any amino acid position ofthe peptide linker;
(ii) adjacent to the C-terminus ofthe second polypeptide (b); and
(iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen;
(d) transforming a host cell with said fourth genetic sequence encoding said single-chain antigen-binding polypeptide of step (c); and (e) expressing said single-chain antigen-binding polypeptide of step (c) in said host, thereby producing a single-chain antigen-binding polypeptide capable of polyalkylene oxide conjugation.
53. The method of claim 52, said first genetic sequence encoding a first polypeptide (a) comprises the antigen binding portion ofthe variable region of an antibody light chain and said second genetic sequence encoding a second polypeptide (b) comprises the antigen binding portion ofthe variable region of an antibody heavy chain.
54. The method of claim 52, wherein the C-terminus of said second polypeptide (b) is the native C-terminus.
55. The method of claim 52, wherein the C-terminus of said second polypeptide (b) comprises a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe second polypeptide are sufficient for the polyalkylene oxide conjugated single-chain antigen-binding polypeptide to be capable of binding an antigen.
56. The method of claim 52, wherein the C-terminus of said second polypeptide (b) comprises an addition of one or plurality of amino acid residue(s), such that the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
57. A multivalent single-chain antigen-binding protein, comprising two or more single-chain antigen-binding polypeptides, each single-chain antigen-binding polypeptide comprising:
(a) a first polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; (b) a second polypeptide comprising the antigen binding portion ofthe variable region of an antibody heavy or light chain; and
(c) a peptide linker linking said first and second polypeptides wherein one ofthe two single-chain antigen-binding polypeptides has at least three consecutive Lys residues which are capable of polyalkylene oxide conjugation and wherein any one of said consecutive Lys residues is located at a position selected from the group consisting of
(i) any amino acid position ofthe peptide linker; (ii) adjacent to the C-terminus ofthe second polypeptide (b); and (iii) combinations thereof, wherein the polyalkylene oxide conjugated single-chain antigen-binding polypeptide is capable of binding an antigen.
58. The multivalent protein of claim 57, wherein said first polypeptide (a) comprises the antigen binding portion ofthe variable region of an antibody light chain and said second polypeptide (b) comprises the antigen binding portion ofthe variable region of an antibody heavy chain.
59. The multivalent protein of claim 57, wherein the C-terminus of said second polypeptide (b) is the native C-terminus.
60. The multivalent protein of claim 57, wherein the C-terminus of said second polypeptide (b) comprises a deletion of one or plurality of amino acid residue(s), such that the remaining N-terminus amino acid residues ofthe second polypeptide are sufficient for the polyalkylene oxide conjugated single-chain antigen-binding polypeptide to be capable of binding an antigen.
61. The multivalent protein of claim 57, wherein the C-terminus of said second polypeptide comprises an addition of one or plurality of amino acid residue(s), such that the polyalkylene oxide conjugated single-chain antigen- binding polypeptide is capable of binding an antigen.
62. The multivalent protein of claim 57, wherein any one of said consecutive Lys residues capable of polyalkylene oxide conjugation is attached to a polyalkylene oxide moiety.
63. The polyalkylene oxide conjugation multivalent protein of claim 62, wherein said polyalkylene oxide conjugated multivalent protein is conjugated to one or plurality of peptide, lipid, nucleic acid, drug, toxin, chelator, boron addend or detectable label molecule(s).
PCT/US1998/008654 1997-04-30 1998-04-30 Polyalkylene oxide-modified single chain polypeptides WO1998048837A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA2288994A CA2288994C (en) 1997-04-30 1998-04-30 Polyalkylene oxide-modified single chain polypeptides
AU72666/98A AU7266698A (en) 1997-04-30 1998-04-30 Polyalkylene oxide-modified single chain polypeptides
JP54734798A JP2002505574A (en) 1997-04-30 1998-04-30 Polyalkylene oxide-modified single-chain polypeptides
EP98920001A EP0979102A4 (en) 1997-04-30 1998-04-30 Polyalkylene oxide-modified single chain polypeptides

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US4444997P 1997-04-30 1997-04-30
US5047297P 1997-06-23 1997-06-23
US6307497P 1997-10-27 1997-10-27
US6734197P 1997-12-02 1997-12-02
US60/067,341 1997-12-02
US60/044,449 1997-12-02
US60/050,472 1997-12-02
US60/063,074 1997-12-02

Publications (2)

Publication Number Publication Date
WO1998048837A1 WO1998048837A1 (en) 1998-11-05
WO1998048837A9 true WO1998048837A9 (en) 1999-04-22

Family

ID=27488916

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US1998/008662 WO1998049198A1 (en) 1997-04-30 1998-04-30 Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
PCT/US1998/008654 WO1998048837A1 (en) 1997-04-30 1998-04-30 Polyalkylene oxide-modified single chain polypeptides

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US1998/008662 WO1998049198A1 (en) 1997-04-30 1998-04-30 Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof

Country Status (6)

Country Link
US (8) US6323322B1 (en)
EP (2) EP0979102A4 (en)
JP (4) JP4187277B2 (en)
AU (2) AU7266898A (en)
CA (2) CA2288994C (en)
WO (2) WO1998049198A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9175083B2 (en) 2004-06-18 2015-11-03 Ambrx, Inc. Antigen-binding polypeptides and their uses
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
US9434778B2 (en) 2014-10-24 2016-09-06 Bristol-Myers Squibb Company Modified FGF-21 polypeptides comprising an internal deletion and uses thereof
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
US9579390B2 (en) 2012-11-12 2017-02-28 Redwood Bioscience, Inc. Compounds and methods for producing a conjugate

Families Citing this family (359)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040009166A1 (en) * 1997-04-30 2004-01-15 Filpula David R. Single chain antigen-binding polypeptides for polymer conjugation
US6323322B1 (en) * 1997-04-30 2001-11-27 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
US6333396B1 (en) 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
AU768295B2 (en) * 1998-11-03 2003-12-04 Centocor Inc. Modified antibodies and antibody fragments with increased duration of activity
US7122632B2 (en) 1999-12-23 2006-10-17 Zymogenetics, Inc. Soluble Interleukin-20 receptor
US6610286B2 (en) 1999-12-23 2003-08-26 Zymogenetics, Inc. Method for treating inflammation using soluble receptors to interleukin-20
EP2133098A1 (en) 2000-01-10 2009-12-16 Maxygen Holdings Ltd G-CSF conjugates
RU2278123C2 (en) 2000-02-11 2006-06-20 Максиджен Холдингз Лтд. Molecules similar to factor vii or viia
US20030027207A1 (en) * 2000-02-29 2003-02-06 Filpula David Ray Anti-platelet binding proteins and polymer conjugates containing the same
US6410271B1 (en) * 2000-06-23 2002-06-25 Genetastix Corporation Generation of highly diverse library of expression vectors via homologous recombination in yeast
US6410246B1 (en) * 2000-06-23 2002-06-25 Genetastix Corporation Highly diverse library of yeast expression vectors
US7118737B2 (en) * 2000-09-08 2006-10-10 Amylin Pharmaceuticals, Inc. Polymer-modified synthetic proteins
AU7338501A (en) * 2000-09-08 2002-03-22 Gryphon Sciences Polymer-modified synthetic proteins
US8110218B2 (en) * 2000-11-30 2012-02-07 The Research Foundation Of State University Of New York Compositions and methods for less immunogenic protein-lipid complexes
AU2002223350A1 (en) * 2000-12-01 2002-06-11 Eleanor N. Fish Cytokine receptor binding peptides
US6979556B2 (en) 2000-12-14 2005-12-27 Genentech, Inc. Separate-cistron contructs for secretion of aglycosylated antibodies from prokaryotes
US7195923B2 (en) * 2001-01-31 2007-03-27 Scripps Laboratories, Inc. Ratiometric determination of glycated protein
IL156059A0 (en) 2001-02-27 2003-12-23 Maxygen Aps NEW INTERFERON beta-LIKE MOLECULES
JP4461210B2 (en) 2001-08-27 2010-05-12 ジェネンテック, インコーポレイテッド Antibody expression system and its construction
US7173003B2 (en) 2001-10-10 2007-02-06 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US7157277B2 (en) 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US7214660B2 (en) 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7795210B2 (en) * 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
AU2004236174B2 (en) 2001-10-10 2011-06-02 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8008252B2 (en) 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
JP4532902B2 (en) 2001-12-17 2010-08-25 ザイモジェネティクス,インコーポレイティド Treatment method for cervical cancer
JP4559081B2 (en) 2002-01-25 2010-10-06 ジーツー セラピーズ リミテッド Anti-C5aR antibody and use thereof
RU2362807C2 (en) 2002-06-21 2009-07-27 Ново Нордиск Хелт Кэр Аг Conjugate of factor vii polypeptide, method of obtaining it, its application and pharmaceutical composition containing it
DE60336555D1 (en) 2002-06-21 2011-05-12 Novo Nordisk Healthcare Ag PEGYLATED GLYCO FORMS OF FACTOR VII
US20050026866A1 (en) * 2002-08-02 2005-02-03 Pawelek John M. Agents and methods for treatment of disease by oligosaccharide targeting agents
US20040067532A1 (en) 2002-08-12 2004-04-08 Genetastix Corporation High throughput generation and affinity maturation of humanized antibody
US7759055B2 (en) * 2002-08-28 2010-07-20 Millipore Corporation Compositions of solution for sequencing reaction clean-up
US8129330B2 (en) 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
US20040062748A1 (en) * 2002-09-30 2004-04-01 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
WO2004061094A1 (en) 2002-12-30 2004-07-22 Gryphon Therapeutics, Inc. Water-soluble thioester and selenoester compounds and methods for making and using the same
US7351688B2 (en) * 2003-02-05 2008-04-01 The Research Foundation Of State University Of New York Compositions and methods for less immunogenic protein formulations
WO2004071420A2 (en) * 2003-02-05 2004-08-26 The Research Foundation Of State University Of New York Compositions and methods for less immunogenic protein formulations
BRPI0408358A (en) * 2003-03-14 2006-03-21 Neose Technologies Inc branched water-soluble polymers and their conjugates
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
EP1624847B1 (en) * 2003-05-09 2012-01-04 BioGeneriX AG Compositions and methods for the preparation of human growth hormone glycosylation mutants
PT1639011E (en) * 2003-06-30 2009-01-20 Domantis Ltd Pegylated single domain antibodies (dab)
WO2005012484A2 (en) * 2003-07-25 2005-02-10 Neose Technologies, Inc. Antibody-toxin conjugates
CA2534565C (en) * 2003-08-05 2012-06-19 The Research Foundation Of State University Of New York Reconstitution medium for protein and peptide formulations
US20050055024A1 (en) * 2003-09-08 2005-03-10 James Anthony H. Orthopaedic implant and screw assembly
WO2005035565A1 (en) 2003-10-10 2005-04-21 Novo Nordisk A/S Il-21 derivatives
ES2428358T3 (en) 2003-10-17 2013-11-07 Novo Nordisk A/S Combination therapy
ATE447587T1 (en) * 2003-11-21 2009-11-15 Zymogenetics Inc ANTI-IL-20 ANTIBODIES AND BINDING PARTNERS AND METHOD FOR USE IN INFLAMMATION
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US20080305992A1 (en) 2003-11-24 2008-12-11 Neose Technologies, Inc. Glycopegylated erythropoietin
US20060040856A1 (en) 2003-12-03 2006-02-23 Neose Technologies, Inc. Glycopegylated factor IX
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US20080220049A1 (en) * 2003-12-05 2008-09-11 Adnexus, A Bristol-Myers Squibb R&D Company Compositions and methods for intraocular delivery of fibronectin scaffold domain proteins
MXPA06006406A (en) 2003-12-05 2007-03-21 Adnexus Therapeutics Inc Inhibitors of type 2 vascular endothelial growth factor receptors.
JP5743368B2 (en) 2004-01-08 2015-07-01 ラショファーム ゲーエムベーハー O-linked glycosylation of peptides
AU2005211362B2 (en) 2004-02-02 2008-03-13 Ambrx, Inc. Modified human interferon polypeptides and their uses
US20080300173A1 (en) 2004-07-13 2008-12-04 Defrees Shawn Branched Peg Remodeling and Glycosylation of Glucagon-Like Peptides-1 [Glp-1]
US20090292110A1 (en) * 2004-07-23 2009-11-26 Defrees Shawn Enzymatic modification of glycopeptides
US20060045866A1 (en) * 2004-09-01 2006-03-02 Chris Chappelow Novel high purity and high molecular weight mPEG alcohol compositions
US8268967B2 (en) 2004-09-10 2012-09-18 Novo Nordisk A/S Glycopegylated interferon α
EP3061461A1 (en) 2004-10-29 2016-08-31 ratiopharm GmbH Remodeling and glycopegylation of fibroblast growth factor (fgf)
WO2006071840A2 (en) 2004-12-22 2006-07-06 Ambrx, Inc. Formulations of human growth hormone comprising a non-naturally encoded amino acid
JP4990792B2 (en) 2004-12-22 2012-08-01 アンブレツクス・インコーポレイテツド Compositions of aminoacyl-tRNA synthetases and uses thereof
MX2007007591A (en) 2004-12-22 2007-07-25 Ambrx Inc Methods for expression and purification of recombinant human growth hormone.
JP2008525473A (en) 2004-12-22 2008-07-17 アンブレツクス・インコーポレイテツド Modified human growth hormone
JP4951527B2 (en) 2005-01-10 2012-06-13 バイオジェネリックス アーゲー GlycoPEGylated granulocyte colony stimulating factor
AU2006208199B2 (en) * 2005-01-25 2012-06-07 Cell Therapeutics, Inc. Conjugates of biologically active proteins having a modified in vivo half-life
WO2006097682A1 (en) * 2005-03-18 2006-09-21 Ucl Business Plc Mechano growth factor peptides and their use
WO2006102659A2 (en) * 2005-03-23 2006-09-28 Nektar Therapeutics Al, Corporation CONJUGATES OF AN hGH MOIETY AND A POLYMER
US20100092505A1 (en) * 2005-04-05 2010-04-15 Elisabetta Bianchi Method for Shielding Functional Sites or Epitopes on Proteins
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
JP5216580B2 (en) 2005-05-25 2013-06-19 ノヴォ ノルディスク アー/エス Glycopegylated factor IX
EP1888098A2 (en) 2005-05-25 2008-02-20 Neose Technologies, Inc. Glycopegylated erythropoietin formulations
US20110003744A1 (en) * 2005-05-25 2011-01-06 Novo Nordisk A/S Glycopegylated Erythropoietin Formulations
JP2008541769A (en) 2005-06-03 2008-11-27 アンブレツクス・インコーポレイテツド Improved human interferon molecules and their use
US8028453B2 (en) * 2005-06-16 2011-10-04 Hold That Thought, Inc. Apparatus and methods for displaying a card
JP5335422B2 (en) 2005-06-17 2013-11-06 ノボ ノルディスク ヘルス ケア アクチェンゲゼルシャフト Selective reduction and derivatization of engineered proteins containing at least one unnatural cysteine
TW200722104A (en) * 2005-06-20 2007-06-16 Pepgen Corp Low-toxicity, long-circulating human interferon-α analogs
US7695710B2 (en) * 2005-06-20 2010-04-13 Pepgen Corporation Antitumor and antiviral combination therapies using low-toxicity, long-circulating human interferon-alpha analogs
NZ565039A (en) * 2005-06-29 2010-04-30 Univ New York State Res Found Compositions and methods for less immunogenic protein-lipid complexes
WO2007021822A2 (en) * 2005-08-09 2007-02-22 The Research Foundation Of State Of University Of New York At Buffalo Compositions and methods of preparation of liposomal microparticulate il-12
US20070105755A1 (en) 2005-10-26 2007-05-10 Neose Technologies, Inc. One pot desialylation and glycopegylation of therapeutic peptides
US20090130776A1 (en) * 2005-09-01 2009-05-21 Canon Kabushiki Kaisha Binding protein molecule
US8168592B2 (en) 2005-10-21 2012-05-01 Amgen Inc. CGRP peptide antagonists and conjugates
US20090048440A1 (en) 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
PT2339014E (en) 2005-11-16 2015-10-13 Ambrx Inc Methods and compositions comprising non-natural amino acids
US7875288B2 (en) * 2006-03-30 2011-01-25 The Research Foundation Of State University Of New York Method for treating blood coagulation disorders
NZ572308A (en) * 2006-03-30 2011-09-30 Univ New York State Res Found Compositions of less immunogenic and long-circulating protein-lipid complexes
EP2010222A1 (en) 2006-03-31 2009-01-07 Baxter International Inc. Pegylated factor viii
US7645860B2 (en) * 2006-03-31 2010-01-12 Baxter Healthcare S.A. Factor VIII polymer conjugates
US7982010B2 (en) * 2006-03-31 2011-07-19 Baxter International Inc. Factor VIII polymer conjugates
US7985839B2 (en) * 2006-03-31 2011-07-26 Baxter International Inc. Factor VIII polymer conjugates
US9101670B2 (en) 2006-04-07 2015-08-11 Nektar Therapeutics Conjugates of an anti-TNF-α antibody
EP2395099A3 (en) 2006-05-02 2012-05-16 Allozyne, Inc. Amino acid substituted molecules
US20080096819A1 (en) * 2006-05-02 2008-04-24 Allozyne, Inc. Amino acid substituted molecules
JP2009537609A (en) 2006-05-24 2009-10-29 ノボ ノルディスク ヘルス ケア アーゲー Extended FIX analogs and derivatives
EP2426150B1 (en) 2006-06-30 2017-10-25 Novo Nordisk A/S Anti-nkg2a antibodies and uses thereof
JP2009544327A (en) 2006-07-21 2009-12-17 ノヴォ ノルディスク アー/エス Glycosylation of peptides with O-linked glycosylation sequences
MX2009002523A (en) 2006-09-08 2009-03-20 Ambrx Inc Modified human plasma polypeptide or fc scaffolds and their uses.
US9133495B2 (en) 2006-09-08 2015-09-15 Ambrx, Inc. Hybrid suppressor tRNA for vertebrate cells
JP2008069073A (en) * 2006-09-12 2008-03-27 Yokohama Tlo Co Ltd Lactoferrin conjugate and its manufacturing method
JP2010503708A (en) * 2006-09-15 2010-02-04 エンゾン ファーマスーティカルズ インコーポレイテッド Targeted polymer prodrugs containing multifunctional linkers
US8367065B2 (en) * 2006-09-15 2013-02-05 Enzon Pharmaceuticals, Inc. Targeted polymeric prodrugs containing multifunctional linkers
US7985783B2 (en) 2006-09-21 2011-07-26 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
JP2010505874A (en) 2006-10-03 2010-02-25 ノヴォ ノルディスク アー/エス Purification method for polypeptide conjugates
EP3156415A1 (en) 2006-11-22 2017-04-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including igf-ir
EP2101821B1 (en) 2006-12-15 2014-08-13 Baxter International Inc. Factor viia- (poly) sialic acid conjugate having prolonged in vivo half-life
CA3001783C (en) * 2007-01-30 2020-09-08 Epivax, Inc. Regulatory t cell epitopes, compositions and uses thereof
EP2111228B1 (en) * 2007-02-02 2011-07-20 Bristol-Myers Squibb Company 10Fn3 domain for use in treating diseases associated with inappropriate angiogenesis
WO2008121563A2 (en) 2007-03-30 2008-10-09 Ambrx, Inc. Modified fgf-21 polypeptides and their uses
ES2406267T3 (en) 2007-04-03 2013-06-06 Biogenerix Ag Treatment methods using glycopegylated G-CSF
US8114630B2 (en) 2007-05-02 2012-02-14 Ambrx, Inc. Modified interferon beta polypeptides and their uses
CN101778859B (en) 2007-06-12 2014-03-26 诺和诺德公司 Improved process for the production of nucleotide sugars
US7968811B2 (en) * 2007-06-29 2011-06-28 Harley-Davidson Motor Company Group, Inc. Integrated ignition and key switch
EP2173380A4 (en) * 2007-07-13 2011-08-31 Abbott Biotech Ltd METHODS AND COMPOSITIONS FOR PULMONARY ADMINISTRATION OF A TNFa INHIBITOR
CA2707840A1 (en) * 2007-08-20 2009-02-26 Allozyne, Inc. Amino acid substituted molecules
EP2190861A4 (en) 2007-08-22 2011-03-30 Univ California Activatable binding polypeptides and methods of identification and use thereof
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
US8877688B2 (en) 2007-09-14 2014-11-04 Adimab, Llc Rationally designed, synthetic antibody libraries and uses therefor
BRPI0816785A2 (en) 2007-09-14 2017-05-02 Adimab Inc rationally designed synthetic antibody libraries, and uses thereof
CA2703267C (en) * 2007-11-15 2015-12-29 Nestec S.A. Production of food products with enhanced in mouth and mental refreshment
US8946148B2 (en) 2007-11-20 2015-02-03 Ambrx, Inc. Modified insulin polypeptides and their uses
US9097667B2 (en) * 2007-12-14 2015-08-04 Biotium, Inc. Fluorescent compounds
CA3102704A1 (en) 2007-12-14 2009-06-25 Novo Nordisk A/S Antibodies against human nkg2d and uses thereof
EP2628753A1 (en) 2008-01-24 2013-08-21 Novo Nordisk A/S Humanized anti-human NKG2A monoclonal antibody
NZ586947A (en) 2008-02-08 2012-11-30 Ambrx Inc Modified leptin polypeptides and their uses
AU2009213141A1 (en) 2008-02-14 2009-08-20 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
BRPI0907532A2 (en) 2008-02-20 2015-07-28 G2 Inflammation Pty Ltd Humanized anti-c5ar antibodies
ES2476690T3 (en) 2008-02-27 2014-07-15 Novo Nordisk A/S Factor VIII conjugated molecules
EP2291399B1 (en) 2008-05-22 2014-06-25 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US10138283B2 (en) 2008-07-23 2018-11-27 Ambrx, Inc. Modified bovine G-CSF polypeptides and their uses
CN102224238B (en) 2008-09-26 2015-06-10 Ambrx公司 Non-natural amino acid replication-dependent microorganisms and vaccines
CN102232085A (en) 2008-09-26 2011-11-02 Ambrx公司 Modified animal erythropoietin polypeptides and their uses
US8927694B2 (en) 2008-11-18 2015-01-06 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
TWI496582B (en) 2008-11-24 2015-08-21 必治妥美雅史谷比公司 Bispecific egfr/igfir binding molecules
AU2009331570B2 (en) 2008-12-22 2013-10-17 Novo Nordisk A/S Antibodies against tissue factor pathway inhibitor
WO2010081173A2 (en) 2009-01-12 2010-07-15 Cytomx Therapeutics, Llc Modified antibody compositions, methods of making and using thereof
DK2379598T3 (en) 2009-01-19 2015-06-15 Innate Pharma Anti-kir3d antibodies
EP2398494A4 (en) * 2009-02-23 2015-10-28 Cytomx Therapeutics Inc Proproteins and methods of use thereof
US8067201B2 (en) * 2009-04-17 2011-11-29 Bristol-Myers Squibb Company Methods for protein refolding
CN102459581B (en) * 2009-06-25 2014-10-29 杜邦营养生物科学有限公司 Protein
WO2011000962A2 (en) 2009-07-03 2011-01-06 Bionor Immuno As Novel therapeutic and diagnostic means
ES2597954T3 (en) * 2009-07-27 2017-01-24 Baxalta GmbH Blood coagulation protein conjugates
CN102573920B (en) 2009-07-27 2015-01-14 利普森技术有限公司 Glycopolysialylation of non-blood coagulation proteins
US8809501B2 (en) 2009-07-27 2014-08-19 Baxter International Inc. Nucleophilic catalysts for oxime linkage
US8642737B2 (en) 2010-07-26 2014-02-04 Baxter International Inc. Nucleophilic catalysts for oxime linkage
HUE028056T2 (en) 2009-07-27 2016-11-28 Baxalta GmbH Blood coagulation protein conjugates
US20120283415A1 (en) 2009-09-10 2012-11-08 Ucb Pharma S.A. Multivalent Antibodies
US8658434B2 (en) * 2009-10-28 2014-02-25 Biotium, Inc. Fluorescent pyrene compounds
GB0920127D0 (en) * 2009-11-17 2009-12-30 Ucb Pharma Sa Antibodies
CN107056929A (en) 2009-12-21 2017-08-18 Ambrx 公司 Porcine somatotropin polypeptide and its purposes by modification
NZ600361A (en) 2009-12-21 2014-06-27 Ambrx Inc Modified bovine somatotropin polypeptides and their uses
TWI513466B (en) 2010-01-20 2015-12-21 Boehringer Ingelheim Int Anticoagulant antidotes
US10080799B2 (en) 2010-02-12 2018-09-25 Arizona Board Of Regents On Behalf Of Arizona State University Methods and compositions related to glycoprotein-immunoglobulin fusions
RS61082B1 (en) 2010-02-26 2020-12-31 Novo Nordisk As Stable antibody containing compositions
AU2011222883B2 (en) 2010-03-05 2016-05-26 Omeros Corporation Chimeric inhibitor molecules of complement activation
TW201138808A (en) 2010-05-03 2011-11-16 Bristol Myers Squibb Co Serum albumin binding molecules
WO2011143274A1 (en) 2010-05-10 2011-11-17 Perseid Therapeutics Polypeptide inhibitors of vla4
WO2011150133A2 (en) 2010-05-26 2011-12-01 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
WO2011147921A1 (en) 2010-05-28 2011-12-01 Novo Nordisk A/S Stable multi-dose compositions comprising an antibody and a preservative
EP2588498B1 (en) 2010-06-29 2018-07-11 Centre National de la Recherche Scientifique (CNRS) Llt-1 antibodies with new functional properties
US9260518B2 (en) 2010-06-30 2016-02-16 Novo Nordisk A/S Antibodies that are capable of specifically binding tissue factor pathway inhibitor
EP4219805A1 (en) 2010-07-16 2023-08-02 Adimab, LLC Antibody libraries
SG187736A1 (en) 2010-08-17 2013-03-28 Ambrx Inc Modified relaxin polypeptides and their uses
TWI480288B (en) 2010-09-23 2015-04-11 Lilly Co Eli Formulations for bovine granulocyte colony stimulating factor and variants thereof
NZ612320A (en) 2010-12-22 2015-06-26 Baxter Healthcare Sa Materials and methods for conjugating a water soluble fatty acid derivative to a protein
MA34978B1 (en) 2011-03-30 2014-03-01 Boehringer Ingelheim Int ANTIDOTES FOR ANTICOAGULANTS
EP2699597B1 (en) 2011-04-21 2016-06-01 Garvan Institute of Medical Research Modified variable domain molecules and methods for producing and using them b
WO2012164021A1 (en) 2011-05-31 2012-12-06 Novo Nordisk A/S Il-21 epitope and il-21 ligands
DK3424953T3 (en) 2011-06-06 2020-11-02 Novo Nordisk As Terapeutiske antistoffer
KR102047248B1 (en) 2011-06-17 2019-11-21 노보 노르디스크 에이/에스 Selective elimination of erosive cells
CA2840552A1 (en) 2011-07-01 2013-01-10 Bayer Intellectual Property Gmbh Relaxin fusion polypeptides and uses thereof
HUE029855T2 (en) 2011-07-05 2017-04-28 Bioasis Technologies Inc P97-antibody conjugates
WO2013011063A1 (en) 2011-07-18 2013-01-24 Novo Nordisk A/S Antagonistic antibodies against oscar
WO2013011061A1 (en) 2011-07-18 2013-01-24 Novo Nordisk A/S Antagonistic antibodies against oscar
WO2013011062A2 (en) 2011-07-18 2013-01-24 Novo Nordisk A/S Oscar antagonists
WO2013011059A1 (en) 2011-07-18 2013-01-24 Novo Nordisk A/S Antagonist antibodies against oscar
WO2013092983A2 (en) 2011-12-23 2013-06-27 Innate Pharma Enzymatic conjugation of polypeptides
CN108530535B (en) 2012-02-15 2021-02-26 诺和诺德股份有限公司 Antibody binding to peptidoglycan-recognizing protein 1
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
LT2814844T (en) 2012-02-15 2017-10-25 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (trem-1)
CA2875624A1 (en) 2012-06-06 2013-12-12 Bionor Immuno As Hiv vaccine
US9738724B2 (en) 2012-06-08 2017-08-22 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9732161B2 (en) 2012-06-26 2017-08-15 Sutro Biopharma, Inc. Modified Fc proteins comprising site-specific non-natural amino acid residues, conjugates of the same, methods of their preparation and methods of their use
WO2014006230A1 (en) 2012-07-06 2014-01-09 Novo Nordisk A/S Il-20 epitopes and il-20 ligands
EP2872894B1 (en) 2012-07-13 2019-04-17 Innate Pharma Screening of conjugated antibodies
CA2880162C (en) 2012-07-31 2023-04-04 Bioasis Technologies, Inc. Dephosphorylated lysosomal storage disease proteins and methods of use thereof
WO2014036520A1 (en) 2012-08-30 2014-03-06 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-erbb3 agents
EP3584255B1 (en) 2012-08-31 2022-02-16 Sutro Biopharma, Inc. Modified amino acids comprising an azido group
EP3564259A3 (en) 2012-11-09 2020-02-12 Innate Pharma Recognition tags for tgase-mediated conjugation
WO2014078733A1 (en) 2012-11-16 2014-05-22 The Regents Of The University Of California Pictet-spengler ligation for protein chemical modification
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
PL2953972T3 (en) 2013-02-05 2021-03-08 Engmab Sàrl Method for the selection of antibodies against bcma
EP2762496A1 (en) 2013-02-05 2014-08-06 EngMab AG Method for the selection of antibodies against BCMA
BR112015022416A2 (en) 2013-03-13 2017-10-24 Bioasis Technologies Inc p97 fragments and their uses
EP2968582B1 (en) 2013-03-15 2020-07-01 Innate Pharma Solid phase tgase-mediated conjugation of antibodies
EP2970499B1 (en) 2013-03-15 2022-08-10 Novo Nordisk A/S Antibodies capable of specifically binding two epitopes on tissue factor pathway inhibitor
EP2789630A1 (en) 2013-04-09 2014-10-15 EngMab AG Bispecific antibodies against CD3e and ROR1
US10071169B2 (en) 2013-06-20 2018-09-11 Innate Pharma Enzymatic conjugation of polypeptides
EP3010548A1 (en) 2013-06-21 2016-04-27 Innate Pharma Enzymatic conjugation of polypeptides
ES2865473T3 (en) 2013-07-10 2021-10-15 Sutro Biopharma Inc Antibodies Comprising Multiple Site-Specific Unnatural Amino Acid Residues, Methods for Their Preparation, and Methods of Use
WO2015007337A1 (en) 2013-07-19 2015-01-22 Bionor Immuno As Method for the vaccination against hiv
US20150093399A1 (en) 2013-08-28 2015-04-02 Bioasis Technologies, Inc. Cns-targeted conjugates having modified fc regions and methods of use thereof
EP3055298B1 (en) 2013-10-11 2020-04-29 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
KR20240034882A (en) 2013-10-15 2024-03-14 씨젠 인크. Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics
TW201605904A (en) 2013-11-07 2016-02-16 諾佛 儂迪克股份有限公司 Novel methods and antibodies for treating coagulapathy
CN105744935B (en) 2013-11-27 2022-09-30 雷德伍德生物科技股份有限公司 Hydrazino-pyrrolo compounds and methods for forming conjugates
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
CN107073119A (en) 2014-02-26 2017-08-18 德克萨斯大学系统董事会 Release for the nitrobenzaldehyde proton of manipulation cell acid poisoning
MY187246A (en) 2014-03-14 2021-09-14 Novartis Ag Antibody molecules to lag-3 and uses thereof
US20170335281A1 (en) 2014-03-15 2017-11-23 Novartis Ag Treatment of cancer using chimeric antigen receptor
EP3572424B1 (en) 2014-03-20 2022-04-20 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type iii domains
JP6702893B2 (en) 2014-06-27 2020-06-03 イナート・ファルマ・ソシエテ・アノニムInnate Pharma Pharma S.A. Multispecific antigen binding protein
CA2952727A1 (en) 2014-06-27 2015-12-30 Innate Pharma Multispecific nkp46 binding proteins
EP3026061A1 (en) 2014-11-26 2016-06-01 Novo Nordisk A/S Site directed mutagenesis of trem-1 antibodies for decreasing viscosity.
EP2975056A1 (en) 2014-07-17 2016-01-20 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
SG10201913702WA (en) 2014-07-17 2020-03-30 Novo Nordisk As Site directed mutagenesis of trem-1 antibodies for decreasing viscosity
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
RU2751660C2 (en) 2014-07-21 2021-07-15 Новартис Аг Treatment of malignant neoplasm using humanized chimeric antigen receptor against bcma
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
JP2017528433A (en) 2014-07-21 2017-09-28 ノバルティス アーゲー Low immunoenhancing dose of mTOR inhibitor and CAR combination
EP3174546B1 (en) 2014-07-31 2019-10-30 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
JP6919118B2 (en) 2014-08-14 2021-08-18 ノバルティス アーゲー Treatment of cancer with GFRα-4 chimeric antigen receptor
JP7084138B2 (en) 2014-08-19 2022-06-14 ノバルティス アーゲー Anti-CD123 Chimeric Antigen Receptor (CAR) for use in cancer treatment
EP3967709A1 (en) 2014-09-17 2022-03-16 Novartis AG Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016055592A1 (en) 2014-10-09 2016-04-14 Engmab Ag Bispecific antibodies against cd3epsilon and ror1
JP6877339B2 (en) 2014-10-14 2021-05-26 ノバルティス アーゲー Antibody molecule against PD-L1 and its use
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
JP2018502084A (en) 2014-12-23 2018-01-25 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Alpha cell regeneration combined with conversion to beta cells
EP4056588A1 (en) 2015-04-08 2022-09-14 Novartis AG Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car)- expressing cell
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
CA2990518A1 (en) 2015-06-23 2016-12-29 Innate Pharma Multispecific nk engager proteins
EP3313876A2 (en) 2015-06-23 2018-05-02 Innate Pharma Multispecific antigen binding proteins
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
EP3964528A1 (en) 2015-07-29 2022-03-09 Novartis AG Combination therapies comprising antibody molecules to lag-3
WO2017021450A1 (en) 2015-08-03 2017-02-09 Engmab Ag Monoclonal antibodies against bcma
ES2781207T3 (en) 2015-09-23 2020-08-31 Bristol Myers Squibb Co Serum albumin-binding fibronectin type iii domains with rapid dissociation rate
SG10202010590UA (en) 2015-12-04 2020-12-30 Seattle Genetics Inc Conjugates of quaternized tubulysin compounds
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
JP2019502695A (en) 2015-12-17 2019-01-31 ノバルティス アーゲー Combination of antibody molecule against PD-1 and C-Met inhibitor and use thereof
MX2018007423A (en) 2015-12-17 2018-11-09 Novartis Ag Antibody molecules to pd-1 and uses thereof.
US20210198368A1 (en) 2016-01-21 2021-07-01 Novartis Ag Multispecific molecules targeting cll-1
JP2019513347A (en) 2016-03-04 2019-05-30 ノバルティス アーゲー Cells expressing multiple chimeric antigen receptor (CAR) molecules and uses thereof
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
MA43835A (en) 2016-03-25 2018-11-28 Seattle Genetics Inc PROCESS FOR THE PREPARATION OF PEGYLATED MEDICINAL PRODUCTS AND THEIR INTERMEDIARIES
DK3443096T3 (en) 2016-04-15 2023-05-22 Novartis Ag COMPOSITIONS AND METHODS FOR SELECTIVE EXPRESSION OF CHIMERIC ANTIGEN RECEPTORS
KR20220103806A (en) 2016-05-18 2022-07-22 베링거 인겔하임 인터내셔날 게엠베하 Anti pd-1 and anti-lag3 antibodies for cancer treatment
US10556936B2 (en) 2016-05-27 2020-02-11 Alk Abelló Immunogenic proteins and fragments thereof from allergenic mites
EP3464375A2 (en) 2016-06-02 2019-04-10 Novartis AG Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
TWI787594B (en) 2016-07-12 2022-12-21 美商凱特製藥公司 Antigen binding molecules and methods of use thereof
JP7219376B2 (en) 2016-07-15 2023-02-08 ノバルティス アーゲー Treatment and prevention of cytokine release syndrome using chimeric antigen receptors in combination with kinase inhibitors
JP2019523301A (en) 2016-07-28 2019-08-22 ノバルティス アーゲー Combination therapy of chimeric antigen receptor and PD-1 inhibitor
AU2017306267A1 (en) 2016-08-01 2019-02-14 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-M2 macrophage molecule
MX2019003886A (en) 2016-10-07 2019-08-05 Novartis Ag Chimeric antigen receptors for the treatment of cancer.
CN110167964B (en) 2016-11-02 2023-12-01 百时美施贵宝公司 Combination of bispecific antibodies and immunopharmaceuticals against BCMA and CD3 for the treatment of multiple myeloma
US11135307B2 (en) 2016-11-23 2021-10-05 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
EP3360898A1 (en) 2017-02-14 2018-08-15 Boehringer Ingelheim International GmbH Bispecific anti-tnf-related apoptosis-inducing ligand receptor 2 and anti-cadherin 17 binding molecules for the treatment of cancer
WO2018115262A1 (en) 2016-12-23 2018-06-28 Innate Pharma Heterodimeric antigen binding proteins
EP4043485A1 (en) 2017-01-26 2022-08-17 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
AU2018219283B2 (en) 2017-02-08 2022-05-19 Bristol-Myers Squibb Company Modified relaxin polypeptides comprising a pharmacokinetic enhancer and uses thereof
US20200048359A1 (en) 2017-02-28 2020-02-13 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
JP2020512312A (en) 2017-03-24 2020-04-23 シアトル ジェネティックス, インコーポレイテッド Process for the preparation of glucuronide drug-linker and its intermediates
US20200179511A1 (en) 2017-04-28 2020-06-11 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
EP3615055A1 (en) 2017-04-28 2020-03-04 Novartis AG Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
US11312783B2 (en) 2017-06-22 2022-04-26 Novartis Ag Antibody molecules to CD73 and uses thereof
US20200223924A1 (en) 2017-06-27 2020-07-16 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
AU2018301393A1 (en) 2017-07-11 2020-02-06 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
CA3070095A1 (en) 2017-07-20 2019-01-24 Novartis Ag Dosage regimens of anti-lag-3 antibodies and uses thereof
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
CA3081602A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
WO2019100052A2 (en) 2017-11-20 2019-05-23 Compass Therapeutics Llc Cd137 antibodies and tumor antigen-targeting antibodies and uses thereof
CA3079363A1 (en) 2017-11-21 2019-05-31 Innate Pharma Multispecific antigen binding proteins
CN111094462A (en) 2017-12-26 2020-05-01 贝克顿·迪金森公司 Deep ultraviolet excitable water-solvated polymer dyes
AU2019215031A1 (en) 2018-01-31 2020-08-20 Novartis Ag Combination therapy using a chimeric antigen receptor
CN112384573A (en) 2018-03-30 2021-02-19 贝克顿·迪金森公司 Water-soluble polymeric dyes containing pending chromophores
CN111936520A (en) 2018-04-02 2020-11-13 百时美施贵宝公司 anti-TREM-1 antibodies and uses thereof
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
US20210047405A1 (en) 2018-04-27 2021-02-18 Novartis Ag Car t cell therapies with enhanced efficacy
EP3569618A1 (en) 2018-05-19 2019-11-20 Boehringer Ingelheim International GmbH Antagonizing cd73 antibody
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
TW202003580A (en) 2018-05-21 2020-01-16 美商坎伯斯治療有限責任公司 Compositions and methods for enhancing the killing of target cells by NK cells
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
EP3806962A1 (en) 2018-06-13 2021-04-21 Novartis AG Bcma chimeric antigen receptors and uses thereof
PE20210418A1 (en) 2018-06-19 2021-03-08 Atarga Llc COMPLEMENT COMPONENT 5 ANTIBODY MOLECULES AND THEIR USES
AR116109A1 (en) 2018-07-10 2021-03-31 Novartis Ag DERIVATIVES OF 3- (5-AMINO-1-OXOISOINDOLIN-2-IL) PIPERIDINE-2,6-DIONA AND USES OF THE SAME
WO2020023300A1 (en) 2018-07-22 2020-01-30 Bioasis Technologies, Inc. Treatment of lymmphatic metastases
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
CN116948006A (en) 2018-09-11 2023-10-27 北京泰德制药股份有限公司 Interleukin-2polypeptide conjugate and use thereof
AU2019361206A1 (en) 2018-10-19 2021-06-03 Ambrx, Inc. Interleukin-10 polypeptide conjugates, dimers thereof, and their uses
KR20210107628A (en) 2018-11-13 2021-09-01 콤파스 테라퓨틱스 엘엘씨 Multispecific binding constructs for checkpoint molecules and uses thereof
GB201818477D0 (en) 2018-11-13 2018-12-26 Emstopa Ltd Tissue plasminogen activator antibodies and method of use thereof
JP2022514315A (en) 2018-12-20 2022-02-10 ノバルティス アーゲー Dosage regimens and drug combinations containing 3- (1-oxoisoindoline-2-yl) piperidine-2,6-dione derivatives
AU2019400980A1 (en) 2018-12-20 2021-06-24 Novartis Ag Pharmaceutical combinations
WO2020168017A1 (en) 2019-02-12 2020-08-20 Ambrx, Inc. Compositions containing, methods and uses of antibody-tlr agonist conjugates
US10871640B2 (en) 2019-02-15 2020-12-22 Perkinelmer Cellular Technologies Germany Gmbh Methods and systems for automated imaging of three-dimensional objects
CN113490528A (en) 2019-02-15 2021-10-08 诺华股份有限公司 3- (1-oxo-5- (piperidine-4-yl) isoindoline-2-yl) piperidine-2, 6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US20220088075A1 (en) 2019-02-22 2022-03-24 The Trustees Of The University Of Pennsylvania Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
MA55519A (en) 2019-03-29 2022-02-09 Atarga Llc ANTI-FGF23 ANTIBODIES
US11208477B2 (en) 2019-04-01 2021-12-28 Novo Nordisk A/S Antibodies and use thereof
WO2021011681A1 (en) 2019-07-15 2021-01-21 Bristol-Myers Squibb Company Antibodies against human trem-1 and uses thereof
US20220372139A1 (en) 2019-07-15 2022-11-24 Bristol-Myers Squibb Company Anti-trem-1 antibodies and uses thereof
JP2022546590A (en) * 2019-09-06 2022-11-04 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Chimeric Antigen Receptors and Related Methods and Compositions for Treatment of Cancer
EP4048285A1 (en) 2019-10-21 2022-08-31 Novartis AG Tim-3 inhibitors and uses thereof
BR112022007376A2 (en) 2019-10-21 2022-07-05 Novartis Ag COMBINATION THERAPIES WITH VENETOCLAX AND TIM-3 INHIBITORS
MX2022006391A (en) 2019-11-26 2022-06-24 Novartis Ag Chimeric antigen receptors binding bcma and cd19 and uses thereof.
IL293834A (en) 2019-12-20 2022-08-01 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
CN113186185B (en) * 2020-01-14 2023-05-26 东北林业大学 Method for efficiently enriching host DNA from mammal feces
KR20220128389A (en) 2020-01-17 2022-09-20 노파르티스 아게 A combination comprising a TIM-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
CN115298322A (en) 2020-01-17 2022-11-04 贝克顿迪金森公司 Methods and compositions for single cell secretogomics
EP4110377A2 (en) 2020-02-27 2023-01-04 Novartis AG Methods of making chimeric antigen receptor-expressing cells
EP4117732A1 (en) 2020-03-11 2023-01-18 Ambrx, Inc. Interleukin-2 polypeptide conjugates and methods of use thereof
US20210355468A1 (en) 2020-05-18 2021-11-18 Bioasis Technologies, Inc. Compositions and methods for treating lewy body dementia
KR20230012559A (en) 2020-05-19 2023-01-26 베링거 인겔하임 인터내셔날 게엠베하 Binding Molecules for Cancer Treatment
TW202210504A (en) 2020-05-29 2022-03-16 德國科隆大學 Neutralizing antibodies against sars-related coronavirus
US20210393787A1 (en) 2020-06-17 2021-12-23 Bioasis Technologies, Inc. Compositions and methods for treating frontotemporal dementia
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
UY39324A (en) 2020-07-16 2022-02-25 Novartis Ag ANTI-BETACELLULIN ANTIBODIES, THEIR FRAGMENTS, MULTISPECIFIC BINDING MOLECULES, EXPRESSION CASSETTES, COMPOSITIONS AND TREATMENT METHODS.
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
EP4192876A1 (en) 2020-08-10 2023-06-14 Innate Pharma Cell surface mica and micb detection using antibodies
CA3190606A1 (en) 2020-08-20 2022-02-24 Ambrx, Inc. Antibody-tlr agonist conjugates, methods and uses thereof
EP4204021A1 (en) 2020-08-31 2023-07-05 Advanced Accelerator Applications International S.A. Method of treating psma-expressing cancers
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
US20220119536A1 (en) 2020-10-21 2022-04-21 Boehringer Ingelheim International Gmbh Agonistic trkb binding molecules for the treatment of eye diseases
IL302277A (en) 2020-10-22 2023-06-01 Janssen Biotech Inc Proteins comprising delta-like ligand 3 (dll3) antigen binding domains and their uses
EP4240765A2 (en) 2020-11-06 2023-09-13 Novartis AG Antibody fc variants
EP4243857A1 (en) 2020-11-13 2023-09-20 Novartis AG Combination therapies with chimeric antigen receptor (car)-expressing cells
EP4284510A1 (en) 2021-01-29 2023-12-06 Novartis AG Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022182872A2 (en) 2021-02-24 2022-09-01 Alladapt Immunotherapeutics, Inc. Compositions and methods for identification of cross-reactive allergenic proteins and treatment of allergies
TW202304990A (en) 2021-03-26 2023-02-01 美商健生生物科技公司 Humanized antibodies against paired helical filament tau and uses thereof
CN117321087A (en) 2021-03-26 2023-12-29 先天制药公司 Multi-specific proteins comprising nkp binding site, cancer antigen binding site fused to cytokine for nk cell engagement
WO2022212899A1 (en) 2021-04-03 2022-10-06 Ambrx, Inc. Anti-her2 antibody-drug conjugates and uses thereof
CA3211948A1 (en) 2021-04-05 2022-10-13 Benjamin Rossi Immunohistochemistry methods and kir3dl2-specific reagents
TW202304979A (en) 2021-04-07 2023-02-01 瑞士商諾華公司 USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
AR125874A1 (en) 2021-05-18 2023-08-23 Novartis Ag COMBINATION THERAPIES
EP4095156A1 (en) 2021-05-28 2022-11-30 Universität zu Köln Neutralizing antibodies against hepatitis c virus
WO2022258678A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp30, a cytokine receptor, a tumour antigen and cd16a
WO2022258691A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkg2d, a cytokine receptor, a tumour antigen and cd16a
CA3227227A1 (en) 2021-06-09 2022-12-15 Laurent Gauthier Multispecific antibodies binding to cd20, nkp46, cd16 and conjugated to il-2
CA3218793A1 (en) 2021-06-09 2022-12-15 Laurent Gauthier Multispecific proteins binding to nkp46, a cytokine receptor, a tumour antigen and cd16a
US20220411533A1 (en) 2021-06-17 2022-12-29 Boehringer Ingelheim International Gmbh Novel Tri-specific Binding Molecules
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
EP4155349A1 (en) 2021-09-24 2023-03-29 Becton, Dickinson and Company Water-soluble yellow green absorbing dyes
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
EP4190810A1 (en) 2021-12-01 2023-06-07 Universität zu Köln Neutralizing antibodies against sars-related coronavirus
WO2023099688A1 (en) 2021-12-01 2023-06-08 Universität Zu Köln Neutralizing antibodies against sars-related coronavirus
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
TW202346355A (en) 2022-03-11 2023-12-01 比利時商健生藥品公司 Multispecific antibodies and uses thereof
WO2023170291A1 (en) 2022-03-11 2023-09-14 Janssen Pharmaceutica Nv Multispecific antibodies and uses thereof
WO2023170295A1 (en) 2022-03-11 2023-09-14 Janssen Pharmaceutica Nv Multispecific antibodies and uses thereof
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2024007016A2 (en) 2022-07-01 2024-01-04 Beckman Coulter, Inc. Novel fluorescent dyes and polymers from dihydrophenanthrene derivatives
US20240052065A1 (en) 2022-07-15 2024-02-15 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
WO2024044327A1 (en) 2022-08-26 2024-02-29 Beckman Coulter, Inc. Dhnt monomers and polymer dyes with modified photophysical properties
WO2024056862A1 (en) 2022-09-15 2024-03-21 Avidicure Ip B.V. Multispecific antigen binding proteins for tumor-targeting of nk cells and use thereof

Family Cites Families (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US5202256A (en) 1984-09-13 1993-04-13 Enzon Labs, Inc. Bioadhesive precursor protein expression vectors
US5202236A (en) 1984-09-13 1993-04-13 Enzon Labs Inc. Method of producing bioadhesive protein
US5049504A (en) * 1986-11-24 1991-09-17 Genex Corporation Bioadhesive coding sequences
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4881175A (en) 1986-09-02 1989-11-14 Genex Corporation Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5260203A (en) * 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US4704692A (en) * 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5869620A (en) 1986-09-02 1999-02-09 Enzon, Inc. Multivalent antigen-binding proteins
US5166320A (en) 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US5122614A (en) 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5358932A (en) * 1989-12-29 1994-10-25 Zymogenetics, Inc. Hybrid protein C
GB9012995D0 (en) * 1990-06-11 1990-08-01 Celltech Ltd Multivalent antigen-binding proteins
US5766897A (en) * 1990-06-21 1998-06-16 Incyte Pharmaceuticals, Inc. Cysteine-pegylated proteins
JPH06506217A (en) 1991-03-18 1994-07-14 エンゾン,インコーポレーテッド Hydrazine-containing conjugates of polypeptides or glycopolypeptides and polymers
US5212075A (en) 1991-04-15 1993-05-18 The Regents Of The University Of California Compositions and methods for introducing effectors to pathogens and cells
US5872222A (en) * 1991-04-19 1999-02-16 Tanox Biosystems, Inc. Conjugates of polymers and antibodies specific for T lymphocytes, and their use as adjuvants
US6787153B1 (en) * 1991-06-28 2004-09-07 Mitsubishi Chemical Corporation Human monoclonal antibody specifically binding to surface antigen of cancer cell membrane
NZ244306A (en) 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5521291A (en) 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
US5981273A (en) * 1991-09-30 1999-11-09 Boehringer Ingelheim Int'l. Gmbh Composition comprising an endosomolytic agent for introducing nucleic acid complexes into higher eucaryotic cells
JPH07501451A (en) 1991-11-25 1995-02-16 エンゾン・インコーポレイテッド Multivalent antigen binding protein
US6025165A (en) * 1991-11-25 2000-02-15 Enzon, Inc. Methods for producing multivalent antigen-binding proteins
CA2129663C (en) * 1992-02-06 2005-07-05 James S. Huston Biosynthetic binding protein for cancer marker
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US6113946A (en) * 1992-04-03 2000-09-05 The Regents Of The University Of California Self-assembling polynucleotide delivery system comprising dendrimer polycations
US6329507B1 (en) 1992-08-21 2001-12-11 The Dow Chemical Company Dimer and multimer forms of single chain polypeptides
WO1994012520A1 (en) 1992-11-20 1994-06-09 Enzon, Inc. Linker for linked fusion polypeptides
US5359030A (en) * 1993-05-10 1994-10-25 Protein Delivery, Inc. Conjugation-stabilized polypeptide compositions, therapeutic delivery and diagnostic formulations comprising same, and method of making and using the same
JPH09504033A (en) 1993-10-20 1997-04-22 エンゾン,インコーポレーテッド 2'- and / or 7-substituted taxoids
US5880131A (en) 1993-10-20 1999-03-09 Enzon, Inc. High molecular weight polymer-based prodrugs
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5844107A (en) * 1994-03-23 1998-12-01 Case Western Reserve University Compacted nucleic acids and their delivery to cells
US5656465A (en) 1994-05-04 1997-08-12 Therion Biologics Corporation Methods of in vivo gene delivery
US5730990A (en) * 1994-06-24 1998-03-24 Enzon, Inc. Non-antigenic amine derived polymers and polymer conjugates
US5888773A (en) * 1994-08-17 1999-03-30 The United States Of America As Represented By The Department Of Health And Human Services Method of producing single-chain Fv molecules
US5670132A (en) 1994-09-20 1997-09-23 Immunomedics, Inc. Modified radioantibody fragments for reduced renal uptake
US5763733A (en) 1994-10-13 1998-06-09 Enzon, Inc. Antigen-binding fusion proteins
JPH09511916A (en) 1994-11-01 1997-12-02 ベルス,ビンフリート Nucleic acid transfer system
AU6255096A (en) 1995-06-07 1996-12-30 Mount Sinai School Of Medicine Of The City University Of New York, The Pegylated modified proteins
US5853723A (en) 1995-09-21 1998-12-29 University Of Utah Research Foundation Targeting of peg antibody conjugates to islet cells
BR9606706A (en) 1995-10-16 1999-04-06 Unilever Nv Bispecific or bivalent antibody fragment analog use process to produce the same
US6090382A (en) * 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
RU2270030C2 (en) * 1996-02-09 2006-02-20 Абботт Байотекнолоджи эЛтиди. METHOD AND OBTAINED HUMAN ANTIBODY OR ITS ANTIGEN-BINDING FRAGMENT FOR INHIBITING HUMAN TNFα ACTIVITY, APPLYING THE OBTAINED HUMAN ANTIBODY OR ITS ANTIGEN-BINDING FRAGMENT AS INGREDIENT FOR PRODUCING MEDICAMENT
GB9625640D0 (en) 1996-12-10 1997-01-29 Celltech Therapeutics Ltd Biological products
CA2281794A1 (en) 1997-02-21 1998-08-27 Genentech, Inc. Antibody fragment-polymer conjugates and humanized anti-il-8 monoclonal antibodies
US6025158A (en) * 1997-02-21 2000-02-15 Genentech, Inc. Nucleic acids encoding humanized anti-IL-8 monoclonal antibodies
US6117980A (en) * 1997-02-21 2000-09-12 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US5830698A (en) * 1997-03-14 1998-11-03 Idec Pharmaceuticals Corporation Method for integrating genes at specific sites in mammalian cells via homologous recombination and vectors for accomplishing the same
US6323322B1 (en) * 1997-04-30 2001-11-27 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
GB9812545D0 (en) 1998-06-10 1998-08-05 Celltech Therapeutics Ltd Biological products
US6333396B1 (en) * 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
US6908963B2 (en) * 2001-10-09 2005-06-21 Nektar Therapeutics Al, Corporation Thioester polymer derivatives and method of modifying the N-terminus of a polypeptide therewith
EP1448602B1 (en) * 2001-11-12 2010-05-26 MERCK PATENT GmbH Modified anti-tnf alpha antibody
US7101978B2 (en) * 2003-01-08 2006-09-05 Applied Molecular Evolution TNF-α binding molecules

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
US9175083B2 (en) 2004-06-18 2015-11-03 Ambrx, Inc. Antigen-binding polypeptides and their uses
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
US9579390B2 (en) 2012-11-12 2017-02-28 Redwood Bioscience, Inc. Compounds and methods for producing a conjugate
US9434778B2 (en) 2014-10-24 2016-09-06 Bristol-Myers Squibb Company Modified FGF-21 polypeptides comprising an internal deletion and uses thereof

Also Published As

Publication number Publication date
US20050008650A1 (en) 2005-01-13
WO1998048837A1 (en) 1998-11-05
WO1998049198A1 (en) 1998-11-05
CA2288994C (en) 2011-07-05
JP2002516610A (en) 2002-06-04
US6824782B2 (en) 2004-11-30
US20020161201A1 (en) 2002-10-31
US6872393B2 (en) 2005-03-29
US6323322B1 (en) 2001-11-27
CA2288992C (en) 2012-06-12
AU7266898A (en) 1998-11-24
US20050048064A1 (en) 2005-03-03
US7632504B2 (en) 2009-12-15
JP2008115193A (en) 2008-05-22
US6743896B2 (en) 2004-06-01
CA2288992A1 (en) 1998-11-05
EP0979102A1 (en) 2000-02-16
JP4187277B2 (en) 2008-11-26
US20050042680A1 (en) 2005-02-24
CA2288994A1 (en) 1998-11-05
US20020061307A1 (en) 2002-05-23
US7150872B2 (en) 2006-12-19
EP0981548A1 (en) 2000-03-01
JP2010051331A (en) 2010-03-11
US20020098192A1 (en) 2002-07-25
EP0979102A4 (en) 2005-11-23
EP0981548A4 (en) 2005-11-23
JP2002505574A (en) 2002-02-19
US6743908B2 (en) 2004-06-01
AU7266698A (en) 1998-11-24
US20020155498A1 (en) 2002-10-24

Similar Documents

Publication Publication Date Title
US6824782B2 (en) Polyalkylene oxide-modified single chain polypeptides
EP3102244B1 (en) Antibody-drug conjugates and immunotoxins
KR0152272B1 (en) Immunoconjugates for cancer diagnosis, and therapy
EA020979B1 (en) PEGYLATED Aβ Fab ANTIBODY FRAGMENTS
CN110740754A (en) kinds of anti-mesothelin antibody and antibody drug conjugate thereof
US20120283418A1 (en) Covalent disulfide-linked diabodies and uses thereof
KR20150036700A (en) Antibody specific for cd22 and methods of use thereof
US20040009166A1 (en) Single chain antigen-binding polypeptides for polymer conjugation
CN113527487A (en) Monoclonal antibody of anti-human B7-H3 and application thereof
US20080125363A1 (en) Polymer-Linked Pseudomonas Exotoxin Immunotoxin

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: C2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: C2

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGES 1/10-10/10, DRAWINGS, REPLACED BY NEW PAGES 1/10-10/10; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref country code: JP

Ref document number: 1998 547347

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref document number: 2288994

Country of ref document: CA

Ref country code: CA

Ref document number: 2288994

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1998920001

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998920001

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642