CA2431600C - Molecules with extended half-lives, compositions and uses thereof - Google Patents

Molecules with extended half-lives, compositions and uses thereof Download PDF

Info

Publication number
CA2431600C
CA2431600C CA2431600A CA2431600A CA2431600C CA 2431600 C CA2431600 C CA 2431600C CA 2431600 A CA2431600 A CA 2431600A CA 2431600 A CA2431600 A CA 2431600A CA 2431600 C CA2431600 C CA 2431600C
Authority
CA
Canada
Prior art keywords
amino acid
substitution
seq
domain
modified molecule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA2431600A
Other languages
French (fr)
Other versions
CA2431600A1 (en
Inventor
William Dall'acqua
Leslie S. Johnson
Elizabeth Sally Ward
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MedImmune LLC
University of Texas System
Original Assignee
MedImmune LLC
University of Texas System
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 MedImmune LLC, University of Texas System filed Critical MedImmune LLC
Priority to CA2766160A priority Critical patent/CA2766160A1/en
Publication of CA2431600A1 publication Critical patent/CA2431600A1/en
Application granted granted Critical
Publication of CA2431600C publication Critical patent/CA2431600C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against Fc-receptors, e.g. CD16, CD32, CD64
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2/00Peptides of undefined number of amino acids; Derivatives thereof
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/53Hinge
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit

Abstract

The present invention provides molecules, including IgGs, non-IgG
immunoglobulins, proteins and non-protein agents, that have increased in vivo half-lives due to the presence of an IgG constant domain, or a portion thereof that binds the FcRn, having one or more amino acid modifications that increase the affinity of the constant domain or fragment for FcRn. Such proteins and molecules with increased half-lives have the advantage that smaller amounts and or less frequent dosing is required in the therapeutic, prophylactic or diagnostic use of such molecules.

Description

MOLECULES WITH EXTENDED HALF-LIVES, COMPOSITIONS AND USES THEREOF

This application claims the benefit of United States provisional application Serial Nos. 60/254,884, filed December 12, 2000, and 60/289,760, filed May 9, 2001. This invention was made, in part, with United States Government support under award number A139167 from the National Institute of Health. The United States Government may have certain rights in the invention.

1. INTRODUCTION
The present invention relates to molecules whose in vivo half-lives are increased by modification of an IgG constant domain, or FcRn (Fc Receptor-neonate) binding domain thereof. Specifically, these molecules have amino acid modifications that increase the affinity of the constant domain or fragment thereof for the FcRn.
Increasing the half-life of therapeutic and diagnostic IgGs and other bioactive molecules using methods of the invention has many benefits including reducing the amount and/or frequency of dosing of these molecules, for example, in vaccines, passive immunotherapy and other therapeutic and prophylactic methods. The invention further relates to fusion proteins containing all or a portion (a FcRn binding portion) of an IgG constant domain having one or more of these amino acid modifications and a non-IgG protein or non protein molecule conjugated to such a modified IgG constant domain, where the presence of the modified IgG
constant domain increases the in vivo half-life of the non-IgG protein or molecule.
2. BACKGROUND OF THE INVENTION
The use of immunoglobulins as therapeutic agents has increased dramatically in recent years and have expanded to different areas of medical treatments.
Such uses include treatment of agammaglobulinemia and hypogammaglobulinemia, as immunosuppressive agents for treating autoimmune diseases and graft-vs.-host (GVH) diseases, the treatment of lymphoid malignancies, and passive immunotherapies for the
3 5 treatment of various systemic and infectious diseases. Also, immunoglobulins are useful as in vivo diagnostic tools, for example, in diagnostic imaging procedures.

One critical issue in these therapies is the persistence of immunoglobulins in the circulation. The rate of immunoglobulin clearance directly affects the amount and frequency of dosage of the immunoglobulin. Increased dosage and frequency of dosage may cause adverse effects in the patient and also increase medical costs.
IgG is the most prevalent immunoglobulin class in humans and other mammals and is utilized in various types of immunotherapies and diagnostic procedures.
The mechanism of IgG catabolism in the circulation has been elucidated through studies related to the transfer of passive immunity from mother to fetus/neonate through the placenta or yolk sac or through colostrum (maternofetal transfer of IgG via transcytosis) in rodents (Brambell, Lancet, ii: 1087-1093, 1966; Rodewald, J Cell Biol., 71:666-670, 1976;
Morris et al., In: Antigen Absorption by the Gut, pp. 3-22, 1978, University Park Press, Baltimore; Jones et al., J. Clin. Invest., 51:2916-2927, 1972).
The involvement of certain receptors in the maternofetal transmission of maternal IgGs was first suggested by Brambell's group in their study on the intestinal absorption of maternal antibodies from ingested milk in newborn rats (Halliday, Proc. R.
Soc. B., 143:408-413, 1955; Halliday, Proc. R. Soc. B., 144:427-430, 1955;
Halliday, Proc.
R. Soc. B., 148:92-103, 1957; Morris, Proc. R. Soc. B., 148:84-91, 1957;
Brambell et al., Proc. R. Soc. B., 149:1-11, 1958; Morris, Proc. R. Soc. B., 160:276-292, 1964). Brambell et al. suggested, based on the observation that heterologous IgGs interfered with the transmission of a specific antibody, that IgG molecules from various species might have sufficiently similar structures or sequences that bind to common receptors (Brambell et al., Proc. R. Soc. B., 149:1-11, 1958).
A high-affinity Fc receptor, FcRn, has been implicated in this transfer mechanism. The FcRn receptor has been isolated from duodenal epithelial brush borders of suckling rats (Rodewald et al., J. Cell Biol., 99:154s-164s, 1984; Simister et al., Eur. J
Immunol., 15:733-738, 1985) and the corresponding gene has been cloned (Simister et al., Nature, 337:184, 1989 and Cold Spring Harbor Symp. Quant. Biol., LIV, 571-580, 1989).
The later clonings of FcRn-encoding genes from mice (Ahouse et al., J
Immunol., 151:6076-6088, 1993) and humans (Story et al., j Exp. Med., 180:2377-2381, 1994) demonstrate high homology of these sequences to the rat FcRn, suggesting a similar mechanism of maternofetal transmission of IgGs involving FcRn in these species.
Meanwhile, a mechanism for IgG catabolism was also proposed by Brambell's group (Brambell et al., Nature, 203:1352-1355, 1964; Brambell, Lancet, ii:1087-1093, 1966). They proposed that a proportion of IgG molecules in the circulation are bound by certain cellular receptors (i.e., FcRn), which are saturable, whereby the IgGs are protected from degradation and eventually recycled into the circulation;
on the other hand, IgGs which are not bound by the receptors are degraded. The proposed mechanism was consistent with the IgG catabolism observed in hypergammaglobulinemic or hypogammaglobulinemic patients. Furthermore, based on his studies as well as others (see, e.g., Spiegelberg et al., J. Exp. Med, 121:323-338, 1965; Edelman et al_, Proc. Natl. AcacL
Sci. USA, 63:78-85, 1969), Brambell also suggested that the mechanisms involved in maternofetal transfer of IgG and catabolism of IgG may be either the same or, at least, very closely related (Brambell, Lancet, ii:1087-1093, 1966). Indeed, it was later reported that a mutation in the Fc-hinge fragment caused concomitant changes in catabolism, matemofetal transfer, neonatal transcytosis, and, particularly, binding to FcRn (Ghetie et al., Immunology Today, 18(12):592-598, 1997).
These observations suggested that portions of the IgG constant domain control IgG metabolism, including the rate of IgG degradation in the serum through interactions with FcRn. Indeed, increased binding affinity for FcRn increased the serum half-life of the molecule (Kim et al., Eur. J Immunol., 24:2429-2434, 1994;
Popov et al., Mol. Immunol., 33:493-502, 1996; Ghetie et al., Eur. J Immunol., 26:690-696, 1996;
Junghans et al., Proc. Natl. Acad. Sci. USA, 93:5512-5516, 1996; Israel et al., ImmunoL, 89:573-578, 1996).
Various site-specific mutagenesis experiments in the Fe region of mouse IgGs have led to identification of certain critical amino acid residues involved in the interaction between IgG and FcRn (Kim et al., Eur. J. ImmunoL, 24:2429-2434, 1994;
Medesan et al., Eur. J ImmunoL, 26:2533, 1996; Medesan et al., J Immunol., 158:2211 -2217, 1997). These studies and sequence comparison studies found that isoleucine at position 253, histidine at position 310, and histidine at position 435 (according to Kabat numbering, Kabat et al., In: Sequences of Proteins of Immunological Interest, US
Department of Health and Human Services, 1991), are highly conserved in human and rodent IgGs, suggesting their importance in IgG-FcRn binding.

Additionally, various publications describe methods for obtaining physiologically active molecules whose half-lives are modified either by introducing an FcRn binding polypeptide into the molecules (WO 97/43316; U.S. Patent No.
5,869,046;
U.S. Patent No. 5,747,035; WO 96/32478; WO 91/14438) or by fusing the molecules with antibodies whose FcRn-binding affinities are preserved but affinities for other Fc receptors have been greatly reduced (WO 99/43713) or fusing with FcRn binding domains of antibodies (WO 00/09560; U.S. Patent No. 4,703,039). However, none of these publications disclose specific mutants in the IgG constant domain that affect half-life.
Prior studies have demonstrated that certain constant domain mutations actually reduce binding to FcRn and, thereby, reduce the IgG in vivo half-life. PCT
publication WO 93/22332 (by Ward et al.) discloses various recombinant mouse IgGs whose in vivo half-lives are reduced by mutations between about residue 253 and about residue 434. Particularly, substitutions of isoleucine at position 253;
histidine at position 310; glutamine at position 311; His at position 433; and asparagine at position 434 were found to reduce IgG half-life.
Modulation of IgG molecules by amino acid substitution, addition, or deletion to increase or reduce affinity for FcRn is also disclosed in WO
98/23289; however, the publication does not list any specific mutants that exhibit either longer or shorter in vivo half-lives.
In fact, only one mutant of mouse IgGI that actually exhibited increased half-life, the triple mutation Thr252 to Ala, Thr254 to Ser, and -Thr256 to Phe, has been identified (WO 97/3463 1).
In view of the pharmaceutical importance of increasing the in vivo half-lives of immunoglobulins and other bioactive molecules, there is a need to develop modified IgGs and FcRn-binding fragments thereof, (particularly modified human IgGs) that confer increased in vivo half-life on immunoglobulins and other bioactive molecules.

3. SUMMARY OF THE INVENTION
The present invention is based upon the inventors' identification of several mutations in the constant domain of a human IgG molecule that increase the affinity of the IgG molecule for the FcRn. In particular, the present inventors have screened libraries of human IgG 1 constant domains with random amino acid mutations introduced into particular regions of the constant domain for increased affinity for FcRn.
Such random mutations were made in the regions of residues 251-256, 285-290, and 308-314, all of which are in CH2 domain, and 385-389 and 428-436, which are in CH3 domain, of human IgGl hinge-Fc regions (residues as depicted in Figure 2 (SEQ ID NO:83 or analogous residues in hinge-Fc regions of other IgG molecules as determined by sequence alignment). As used herein, all residues of the IgG constant domain are numbered according to Kabat et al. (Sequences of Proteins of Immunological Interest, U.S.
Department of Health and Human Services, 1991) and as presented in Figure 2 (SEQ
ID NO:83), and include corresponding residues in other IgG constant domains as
-4-determined by sequence alignment. The in vivo half-life, or persistence in serum or other tissues of a subject, of antibodies, and other therapeutic agents and other bioactive molecules is an important clinical parameter which determines the amount and frequency of antibody (or any other pharmaceutical molecule) administration. Accordingly, such molecules, including antibodies, with increased half-life are of significant pharmaceutical importance.
Thus, the present invention relates to a modified molecule (preferably a protein, but may be a non-protein agent) that has an increased in vivo half-life by virtue of the presence of a modified IgG constant domain, or FcRn-binding portion thereof (preferably the Fe or hinge-Fe domain) (preferably from a human IgG) wherein the IgG
constant domain, or fragment thereof, is modified (e.g., by amino acid substitution, deletion or insertion) to increase the affinity for the FcRn. In a particular embodiment, the present invention relates to modified IgGs, whose in vivo half-lives are extended by the modification of amino acid residues identified to be involved in the interaction of the hinge-Fc domain with the FcRn receptor. Preferably, the constant domain or fragment thereof has higher affinity for FcRn at pH 6.0 than at pH 7.4. Such modifications may also alter (i.e., increase or decrease) the bioavailability (e.g., transport to mucosal surfaces, or other target tissues) of the molecules. The invention also relates to other types of immunoglobulins or fragments thereof (i. e., non-IgG immunoglobulins), non-immunoglobulin proteins and non-protein agents that are fused or conjugated to, or engineered to contain, an IgG constant domain, or FcRn-binding fragment thereof, having one or more such amino acid modifications.
In preferred embodiments, the present invention provides molecules, particularly, immunoglobulins whose in vivo half-lives are extended by the presence of an IgG constant domain, or FcRn binding fragment thereof (preferably, Fc or hinge-Fc domain), that has modifications of one or more of amino acid residues 251-256, 285-290, 308-314, 385-389, and 428-436 that increase the affinity of the constant domains or fragments thereof for FeRn. In certain embodiments, these modifications preferably exclude residues 252, 254, and 256, in particular when the IgG constant domain or fragment thereof, is murine. In particular embodiments, the modification is at one or more surface-exposed residues, and the modification is a substitution with a residue of similar charge, polarity or hydrophobicity to the residue being substituted. In preferred embodiments, the modified IgG constant domain, or fragment thereof, binds with higher affinity to FeRn at pH 6.0 than at pH 7.4. In a preferred embodiment, the constant domain, or fragment thereof, is modified by substitution of one or more of amino acid residues 251-256, 285-
-5-290, 308-314, 385-389, and 428-436 that increase the affinity of the constant domain or FcRn-binding fragments thereof for FcRn. In certain embodiments, substitutions of residue 252 with leucine, residue 254 with serine, and/or residue 256 with phenylalanine are excluded, particularly when the constant domain or fragment thereof is derived from a mouse IgG.
In specific embodiments, the invention provides immunoglobulins or other bioactive molecules that contain an IgGi constant domain, or FcRn-binding fragment thereof (preferably Fc or hinge-Fe domain) (preferably human), having amino acid modifications at one or more of position 308, 309, 311, 312, and 314, more specifically, having substitutions at one or more of positions 308, 309, 311, 312 and 314 with threonine, proline, serine, aspartic acid and leucine respectively. In another embodiment, residues at one or more of positions 308, 309, and 311 are substituted with isoleucine, proline, and glutamic acid, respectively. In yet another embodiment, residues at one or more of positions 308, 309, 311, 312, and 314, are substituted with threonine, proline, serine, aspartic acid, and leucine, respectively. The invention further relates to combinations of these amino acid substitutions.
Furthermore, the invention provides immunoglobulins or other bioactive molecules that contain an IgG1 constant domain, or FcRn-binding fragment thereof (preferably, Fc or hinge-Fc domain) (preferably human), having amino acid modifications at one or more of positions 251, 252, 254, 255, and 256, more specifically, having substitutions at one or more of these positions. In specific embodiments, residue 251 is substituted with leucine or arginine, residue 252 is substituted with tyrosine, phenylalanine, serine, tryptophan or threonine, residue 254 is substituted with threonine or serine, residue 255 is substituted with leucine, glycine, isoleucine or arginine, and/or residue 256 is substituted with serine, arginine, glutainine, glutamic acid, aspartic acid, alanine, asparagine or threonine. In a more specific embodiment, residue 251 is substituted with leucine, residue 252 is substituted with tyrosine, residue 254 is substituted with threonine or serine, and/or residue 255 is substituted with arginine. In yet another specific embodiment, residue 252 is substituted with phenylalanine and/or residue 256 is substituted with aspartic acid. In a preferred embodiment, residue 251 is substituted with leucine, residue 252 is substituted with tyrosine, residue 254 is substituted with threonine or serine, and/or residue 255 is substituted with arginine. The invention further relates to any combination of these substitutions.
Furthermore, the invention provides immunoglobulins or other bioactive molecules that contain an IgG1 constant domain, or FcRn-binding fragment thereof
-6-(preferably, Fc or hinge-Fc domain) (preferably human), having amino acid modifications at one or more of positions 428, 433, 434, and 436, more specifically, having substitutions at one or more of these positions. In specific embodiments, residue 428 is substituted with methionine, threonine, leucine, phenylalanine, or serine, residue 433 is substituted with lysine, arginine, serine, isoleucine, proline, glutamine, or histidine, residue 434 is substituted with phenylalanine, tyrosine, or histidine, and/or residue 436 is substituted with histidine, asparagine, arginine, threonine, lysine, methionine, or threonine.
In a more specific embodiment, residues at one or more positions 433, 434, and 436 are substituted with lysine, phenylalanine, and histidine, respectively. In a preferred embodiment, residue 428 is substituted with methionine and/or residue 434 is substituted with tyrosine.
Furthermore, the invention provides immunoglobulins or other bioactive molecules that contain an IgGl constant domain, or FcRn-binding fragment thereof (preferably, Fc or hinge-Fc domain) (preferably human), having amino acid modifications at one or more positions 385, 386, 387, and 389, more specifically, having substitutions at one or more of these positions. In specific embodiments, residue 385 is substituted with arginine, aspartic acid, serine, threonine, histidine, lysine, or alanine, residue 386 is substituted with threonine, proline, aspartic acid, serine, lysine, arginine, isoleucine, or methionine, residue 387 is substituted with arginine, histidine, serine, threonine, alanine, or proline and/or residue 389 is substituted with proline or serine. In more specific embodiments, residues at one or more positions 385, 386, 387, and 389 are substituted with arginine, threonine, arginine, and proline, respectively. In yet another specific embodiment, residues at one or more positions 385, 386, and 389 are substituted with aspartic acid, proline, and serine, respectively.
Molecules of the invention include any combination of the above-described substitutions at one or more of residues 251, 252, 254, 255, 256, 308, 309, 311, 312, 385, 386, 387, 389, 428, 433, 434, and/or436. In a preferred embodiment, the molecule of the invention contains a Fe region, or FcRn-binding domain thereof, having one or more of the following substitutions: leucine at residue 251, tyrosine at residue 252, threonine or serine at residue 254, arginine at residue 255, threonine at residue 308, proline at residue 309, serine at residue 311, aspartic acid at residue 312, leucine at residue 314, arginine at residue 385, threonine at residue 386, arginine at residue 387, proline at residue 389, methionine at residue 428, and/or tyrosine at residue 434.
Included within the invention are pharmaceutical compositions and methods of prophylaxis and therapy using modified immunoglobulins, proteins and other bioactive molecules of the invention having extended half-lives. Also included are methods of
-7-diagnosis using modified immunoglobulins, proteins and other bioactive molecules of the invention having extended half-lives. In a specific embodiment, the invention provides an anti-respiratory syncytial virus (RSV) antibody useful to treat or prevent RSV
infection, such as SYNAGIS (see U.S. Patent No. 5,824,307 and Johnson et al., J.
Infectious Disease 176:1215-1224, 1997), and other anti-RSV antibodies, including variants of SYNAGIS
(see United States patent Application Serial No., 09/724,396, filed November 28, 2000, United States patent Application Serial No. 09/724,531, filed November 28, 2000, United States patent No.6,855,493 and United States patent No. 6,818,216, all entitled "Methods of Administering/Dosing Anti-RSV Antibodies for Prophylaxis and Treatment,"
all by Young et al., particularly the sequences of heavy and light chain variable domains and CDRs of anti-RSV antibodies disclosed therein), which has one or more amino acid modifications in the constant domain that increase the affinity of the antibody for FcRn and that has an increased in vivo half-life (see also, Section 5.1 infra).

3.1 DEFINITIONS
The term "IgG Fc region" as used herein refers to the portion of an IgG
molecule that correlates to a crystallizable fragment obtained by papain digestion of an IgG
molecule. The Fe region consists of the C-terminal half of the two heavy chains of an IgG
molecule that are linked by disulfide bonds. It has no antigen binding activity but contains the carbohydrate moiety and the binding sites for complement and Fc receptors, including the FcRn receptor (see below). The Fc fragment contains the entire second constant domain CH2 (residues 231-340 of human IgGI, according to the Kabat numbering system) (e.g., SEQ ID NO:80) and the third constant domain CH3 (residues 341-447) (e.g., SEQ
NO:81).
The term "IgG hinge-Fc region" or "hinge-Fc fragment" as used herein refers to a region of an IgG molecule consisting of the Fc region (residues 231-447) and a hinge region (residues 216-230; e.g., SEQ ID NO:82) extending from the N-terminus of the Fc region. An example of the amino acid sequence of the human IgGI hinge-Fc region is SEQ
ID NO:83.
The term "constant domain" refers to the portion of an immunoglobulin molecule having a more conserved amino acid sequence relative to the other portion of the immunoglobulin, the variable domain, which contains the antigen binding site.
The
-8-
9 PCT/US01/48432 constant domain contains the CH1, CH2 and CH3 domains of the heavy chain and the CHL
domain of the light chain.
The term "FcRn receptor" or "FcRn" as used herein refers to an Fc receptor ("n" indicates neonatal) which is known to be involved in transfer of maternal IgGs to a fetus through the human or primate placenta, or yolk sac (rabbits) and to a neonate from the colostrum through the small intestine. It is also known that FcRn is involved in the maintenance of constant serum IgG levels by binding the IgG molecules and recycling them into the serum. The binding of FcRn to IgG molecules is strictly pH-dependent with optimum binding at pH 6Ø FcRn comprises a heterodimer of two polypeptides, whose molecular weights are approximately 50 kD and 15 kD, respectively. The extracellular domains of the 50 kD polypeptide are related to major histocompatibility complex (MHC) class I a-chains and the 15 kD polypeptide was shown to be the non-polymorphic microglobulin ((32-m). In addition to placenta and neonatal intestine, FcRn is also expressed in various tissues across species as well as various types of endothelial cell lines. It is also expressed in human adult vascular endothelium, muscle vasculature and hepatic sinusoids and it is suggested that the endothelial cells may be most responsible for the maintenance of serum IgG levels in humans and mice. The amino acid sequences of human FcRn and marine FcRn are indicated by SEQ ID NO:84 and SEQ ID NO:85, respectively.
Homologs of these sequences having FcRn activity are also included.
The terin "in vivo half-life" as used herein refers to a biological half-life of a particular type of IgG molecule or its fragments containing FcRn-binding sites in the circulation of a given animal and is represented by a time required for half the quantity administered in the animal to be cleared from the circulation and/or other tissues in the animal. When a clearance curve of a given IgG is constructed as a function of time, the curve is usually biphasic with a rapid a-phase which represents an equilibration of the injected IgG molecules between the intra- and extra-vascular space and which is, in part, determined by the size of molecules, and a longer (3-phase which represents the catabolism of the IgG molecules in the intravascular space. The term "in vivo half-life"
practically corresponds to the half life of the IgG molecules in the f3-phase.
An "isolated" or "purified" antibody or fusion protein is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material"
includes preparations of an antibody or a fusion protein in which the antibody or the fusion protein is separated from cellular components of the cells from which it is isolated or recombinantly produced. Thus, an antibody or a fusion protein that is substantially free of cellular material includes preparations of antibody or fusion protein having less than about 30%, 20%, 10%, or 5% (by dry weight) of contaminating protein. When the antibody or the fusion protein is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, 10%, or 5% of the volume of the protein preparation. When the antibody or the fusion protein is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly such preparations of the antibody or the fusion protein have less than about 30%, 20%, 10%, 5% (by dry weight) of chemical precursors or compounds other than the antibody or antibody fragment of interest. In a preferred embodiment of the present invention, antibodies are isolated or purified. In another preferred embodiment of the invention, fusion proteins are isolated or purified.
An "isolated" nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid molecule.
Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized. An "isolated" nucleic acid molecule does not include cDNA
molecules within a cDNA library. In a preferred embodiment of the invention, nucleic acid molecules encoding antibodies are isolated or purified. In another preferred embodiment of the invention, nucleic acid molecules encoding fusion proteins are isolated or purified.
The term "host cell" as used herein refers to the particular subject cell transfected with a nucleic acid molecule or infected with phagemid or bacteriophage and the progeny or potential progeny of such a cell. Progeny of such a cell may not be identical to the parent cell transfected with the nucleic acid molecule due to mutations or environmental influences that may occur in succeeding generations or integration of the nucleic acid molecule into the host cell genome.
The names of amino acids referred to herein are abbreviated either with three-letter or one-letter symbols.
To determine the percent identity of two amino acid sequences or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino acid or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are
-10-then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., %
identity =
number of identical overlapping positions/total number of positions x 100%).
In one embodiment, the two sequences are the same length.
The determination of percent identity between two sequences can also be accomplished using a mathematical algorithm. A preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, 1990, Proc. Natl. Acad Sci. USA. 87:2264-2268, modified as in Karlin and Altschul, 1993, Proc. Natl. Acad. Sci. U.S.A. 90:5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al., 1990, J. Mol.
Biol. 215:403. BLAST nucleotide searches can be performed with the NBLAST
nucleotide program parameters set, e.g., for score=100, wordlength=12to obtain nucleotide sequences homologous to a nucleic acid molecule of the present invention. BLAST protein searches can can be performed with the XBLAST program parameters set, e.g, to score-50, wordlength=3 to obtain amino acid sequences homologous to a protein molecule of the present invention. To obtain gapped alignments for comparison purposes, Gapped BLAST
can be utilized as described in Altschul et al., 1997, Nucleic Acids Res.
25:3389-3402.
Alternatively, PSI-BLAST can be used to perform an iterated search which detects distant relationships between molecules (Id). When utilizing BLAST, Gapped BLAST, and PSI-BLAST programs, the default parameters of the respective programs (e.g., of XBLAST
and NBLAST) can be used (see, e.g., the ncbi.nih.gov website). Another preferred, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, 1988, CABIOS 4:11-17. Such an algorithm is incorporated in the ALIGN program (version 2.0) which is part of the GCG
sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM 120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used.
The percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically only exact matches are counted.

-ll-4. DESCRIPTION OF THE FIGURES
FIG. 1 shows the structure of the IgG hinge-Fc region indicating the locations of the residues identified to be involved in the interaction with the FcRn receptor (Ghetie et al., Immunology Today, 18(12):592-598, 1997).
FIG. 2 shows the amino acid sequence of the human IgGI hinge-Fc region (SEQ ID NO:83) containing a hinge region (SEQ ID NO:82), CH2 domain (SEQ ID
NO:80), and CH3 domain (SEQ ID NO:81).
FIGS. 3 (A and B) show the amino acid sequences of (A) human FcRn (SEQ
ID NO:84) and (B) mouse FcRn (SEQ IDNO:85), respectively.
FIG. 4 shows the amino acid sequence of the human IgGI hinge-Fc region (SEQ ID NO:83), in which wild-type residues which are mutated by amino acid substitutions are indicated in underlined bold-face.
FIG. 5 shows a schematic diagram of panning process for the phage-displayed modified hinge-Fc library.
FIG. 6 shows a summary of the occurrence of selected mutant residues at the variant positions in the libraries screened.
FIGS. 7 (A-D). (A) shows the binding of murine FcRn to immmobilized IgGI having M252Y/S254T/T256E substitutions. Murine FcRn was injected at 10 different concentrations ranging from 1nM to 556 nM over a surface on which 4000 resonance units (RU) of IgGI had been coupled. After equilibrium was reached, residual bound protein was eluted with a pulse of PBS, pH 7.4. (B) shows the binding of human FcRn to immobilized IgGl/M252Y/S254T/T256E. Human FcRn was injected at 8 different concentrations ranging from 71 nM to 2.86 M over a surface on which 1000 RU
of IgGI
had been coupled. After equilibrium was reached, residual bound protein was eluted with a pulse of PBS, pH 7.4. (C) and (D) show scatchard analyses of the data in (A) and (B), respectively, after correction for nonspecific binding. Rq is the corrected equilibrium response at a given concentration C. The plots are linear with correlation coefficients of 0.97 and 0.998, respectively. The apparent Kd are 24 nM and 225 nM, respectively.
FIGS. 8 (A-H). (A)-(D) show the results from BlAcore analysis of the binding of murine FeRn at pH 6.0 and pH 7.4 to (A) wild type human IgGI, (B) M252Y/S254T/T256E, (C) H433K/N434F/Y436H, and (D) G385D/G386P/N389S, respectively, after correction for nonspecific binding. Murine FcRn was injected at a concentration of 1.1 m over a surface on which 1000 RU of wild type IgGI, 1000 RU of M252Y/S254T/T256E, 955 RU of H433K/N434F/Y436H, and 939 RU of G385D/Q386P/N389S had been coupled. (E)-(H) show the results from BlAcore analysis of the binding of human FcRn at pH 6.0 and pH 7.4 to (E) wild type human IgGI, (F) M252Y/S254T/T256E, (G) H433K/N434F/Y436H, and (H) G385D/Q386P/N389S, respectively, after correction for nonspecific binding. Human FcRn was injected at a concentration of 1.4 .tm over a surface on which 1000 RU of wild type IgGI, 1000 RU of M252Y/S254T/T256E, 955 RU of H433K/N434F/Y436H, and 939 RU of G385D/Q386P/N389S had been coupled.
FIG. 9 shows the space-filling model of the surface of the Fc fragment of a human IgGI based upon the human IgGI structure of Deisenhofer, 1981, Biochemistry 20:2361-2370. Residues are color-coded according to the gain of free energy of stabilization of the Fc-FcRn complex: dark grey, substitutions at these positions (M252, S254, T256, H433, N434, and Y436) were found to increase affinity by a factor of at least 2.5 times in the Fc/human FcRn interaction and of at least 5 times in the Fc/mouse FcRn interaction; light grey, substitutions at those positions (G385, G386, P387, N389, V308, L309, and Q311) were found to increase affinity by a factor of less than 2 times in both the Fc-human FcRn and Fc-mouse FcRn interaction. The figure was drawn using Swiss pdb viewer (Guex and Peitsch, 1997, Electrophoresis 18:2714-2723).
FIG. 10 shows the changes in serum concentration ([Mab] ng/ml) over time (in days) of antibody having a wild type constant domain (SYNAGIS ) (open squares), or constant domains with the following mutations: M252Y/S254T/T256E (open circles), G385D/Q386P/N389S (solid squares), and H433K/N434F/Y436H (solid circles).
Antibody concentration was determined using anti-human IgG ELISA.

5. DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to molecules, particularly proteins, more particularly immunoglobulins, that have an increased in vivo half-life and comprise an IgG
constant domain, or fragment thereof that binds to an FcRn (preferably a Fc or hinge-Fc domain), that contains one or more amino acid modifications relative to a wild type IgG
constant domain which modifications increase the affinity of the IgG constant domain, or fragment thereof, for the FcRn. In a preferred embodiment, the invention particularly relates to the modification of human or humanized IgGs and other bioactive molecules containing FcRn-binding portions of human IgGs, which have particular use in human therapy, prophylaxis and diagnosis.

5.1 MOLECULES WITH INCREASED IN VIVO
HALF-LIVES
The present invention is based upon identification of amino acid modifications in particular portions of the IgG constant domain that interact with the FeRn, which modifications increase the affinity of the IgG, or fragment thereof, for the FcRn.
Accordingly, the invention relates to molecules, preferably proteins, more preferably immunoglobulins, that comprise an IgG constant domain, or FcRn binding fragment thereof (preferably a Fc or hinge-Fc domain fragment), having one or more amino acid modifications (i.e., substitutions, insertions or deletions) in one or more regions that interact with the FcRn, which modifications increase the affinity of the IgG or fragment thereof, for the FcRn, and also increase the in vivo half-life of the molecule. In preferred embodiments, the one or more amino acid modifications are made in one or more of residues 251-256, 285-290, 308-314, 385-389, and 428-436 of the IgG hinge-Fc region (for example, as in the human IgG1 hinge-Fc region depicted in Figure 4, SEQ ID NO:83), or analogous residues thereof, as determined by amino acid sequence alignment, in other IgG hinge-Fc regions. In a preferred embodiment, the amino acid modifications are made in a human IgG
constant domain, or FcRn-binding domain thereof. In a certain embodiment, the modifications are not made at residues 252, 254, or 256 (i.e., all are made at one or more of residues 251, 253, 255, 285-290, 308-314, 385-389, or 428-436) of the IgG constant domain. In a more preferred embodiment, the amino acid modifications are not the substitution with leucine at residue 252, with serine at 254, and/or with phenylalanine at position 256. In particular, in preferred embodiments, such modifications are not made when the IgG constant domain, hinge-Fc domain, hinge-Fc domain or other FcRn-binding fragment thereof is derived from a mouse.
The amino acid modifications may be any modification, preferably at one or more of residues 251-256, 285-290, 308-314, 385-389, and 428-436, that increases the in vivo half-life of the IgG constant domain, or FcRn-binding fragment thereof (e.g., Fc or hinge-Fc domain), and any molecule attached thereto, and increases the affinity of the IgG, or fragment thereof, for FcRn. Preferably, the one or more modifications also result in a higher binding affinity of the constant domain, or FcRn-binding fragment thereof, for FeRn at pH 6.0 than at pH 7.4. In other embodiments, the modifications alter (i.e., increase or decrease) bioavailability of the molecule, in particular, alters (i.e., increases or decreases) transport (or concentration or half-life) of the molecule to mucosal surfaces (e.g., of the lungs) or other portions of a target tissue. In a preferred embodiment, the amino acid modifications alter (preferably, increase) transport or concentration or half-life of the molecule to the lungs. In other embodiments, the amino acid modifications alter (preferably, increase) transport (or concentration or half-life) of the molecule to the heart, pancreas, liver, kidney, bladder, stomach, large or small intestine, respiratory tract, lymph nodes, nervous tissue (central and/or peripheral nervous tissue), muscle, epidermis, bone, cartilage, joints, blood vessels, bone marrow, prostate, ovary, uterine, tumor or cancer tissue, etc. In a preferred embodiment, the amino acid modifications do not abolish, or, more preferably, do not alter, other immune effector or receptor binding functions of the constant domain, for example, but not limited to complement fixation, ADCC and binding to FcyRI, FcyRII, and FcyRIII, as can be determined by methods well-known and routine in the art. In another preferred embodiment, the modified FcRn binding fragment of the constant domain does not contain sequences that mediate immune effector functions or other receptor binding. Such fragments may be particularly useful for conjugation to a non-IgG or non-immunoglobulin molecule to increase the in vivo half-life thereof.
In yet another embodiment, the effector functions are selectively altered (e.g., to reduce or increase effector functions).
In preferred embodiments, the amino acid modifications are substitutions at one or more of residues 308, 309, 311, 312 and 314, particularly a substitution with threonine at position 308, proline at position 309, serine at position 311, aspartic acid at position 312, and/or leucine at position 314. Alternatively, the modification is the substitution with an isoleucine at position 308, proline at position 309, and/or a glutamic acid at position 311. In yet another embodiment, residues at one or more of positions 308, 309, 311, 312, and 314, are substituted with threonine, proline, leucine, alanine, and alanine, respectively. Accordingly, in certain embodiments the residue at position 308 is substituted with threonine or isoleucine, the residue at position 309 is substituted with proline, the residue at position 311 is substituted with serine, glutamic acid or leucine, the residue at position 312 is substituted with alanine, and/or the residue at position 314 is substituted with leucine or alanine. In a preferred embodiment, the substitution is a threonine at position 308, a proline at position 309, a serine at position 311, an aspartic acid at position 312, and/or a leucine at position 314.
In preferred embodiments, the amino acid modifications are substitutions at one or more of residues 251, 252, 254, 255, and 256. In specific embodiments, residue 251 is substituted with leucine or arginine, residue 252 is substituted with tyrosine, phenylalanine, serine, tryptophan or threonine, residue 254 is substituted with threonine or serine, residue 255 is substituted with arginine, leucine, glycine, or isoleucine, and/or residue 256 is substituted with serine, arginine, glutamine, glutamic acid, aspartic acid, alanine, asparagine or threonine. In a more specific embodiment, residue 251 is substituted with leucine, residue 252 is substituted with tyrosine, residue 254 is substituted with threonine or serine, residue 255 is substituted with arginine, and/or residue 256 is substituted with glutainic acid.
In preferred embodiments, the amino acid modifications are substitutions at one or more of residues 428, 433, 434, and 436. In specific embodiments, residue 428 is substituted with threonine, methionine, leucine, phenylalanine, or serine, residue 433 is substituted with lysine, arginine, serine, isoleucine, proline, glutamine or histidine, residue 434 is substituted with phenylalanine, tyrosine, or histidine, and/or residue 436 is substituted with histidine, asparagine, arginine, threonine, lysine, or methionine. In a more specific embodiment, residues at position 428 and/or 434 are substituted with methionine, and/or histidine respectively.
In preferred embodiments, the amino acid modifications are substitutions at one or more of residues 385, 386, 387, and 389, more specifically, having substitutions at one or more of these positions. In specific embodiments, residue 385 is substituted with arginine, aspartic acid, serine, threonine, histidine, lysine, alanine or glycine, residue 386 is substituted with threonine, proline, aspartic acid, serine, lysine, arginine, isoleucine, or methionine, residue 387 is substituted with arginine, proline, histidine, serine, threonine, or alanine, and/or residue 389 is substituted with proline, serine or asparagine.
In more specific embodiments, residues at one or more positions 385, 386, 387, and 389 are substituted with arginine, threonine, arginine, and proline, respectively. In yet another specific embodiment, residues at one or more positions 385, 386, and 389 are substituted with aspartic acid, proline, and serine, respectively.
In particular embodiments, amino acid modifications are made at one or a combination of residues 251, 252, 254, 255, 256, 308, 309, 311, 312, 314, 385, 386, 387, 389, 428, 433, 434, and/or 436, particularly where the modifications are one or more of the amino acid substitutions described immediately above for these residues.
In a preferred embodiment, the molecule of the invention contains a Fe region, or FcRn-binding domain thereof, having one or more of the following substitutions:
leucine at residue 251, tyrosine at residue 252, threonine or serine at residue 254, arginine at residue 255, threonine at residue 308, proline at residue 309, serine at residue 311, aspartic acid at residue 312, leucine at residue 314, arginine at residue 385, threonine at residue 386, arginine at residue 387, proline at residue 389, methionine at residue 428, and/or tyrosine at residue 434.

In a preferred embodiment, the FcRn binding domain has a substitution at 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16 or all 18 of residues 251, 252, 254, 255, 256, 308, 309, 311, 312, 314, 385, 386, 387, 389, 428, 433, 434, and/or 436.
Amino acid modifications can be made by any method known in the art and many such methods are well known and routine for the skilled artisan. For example, but not by way of limitation, amino acid substitutions, deletions and insertions may be accomplished using any well-known PCR-based technique. Amino acid substitutions may be made by site-directed mutagenesis (see, for example, Zoller and Smith, Nucl. Acids Res.
10:6487-6500, 1982; Kunkel, Proc. Natl. Acad. Sci USA 82:488, 1985). Mutants that result in increased affinity for FeRn and increased in vivo half-life may readily be screened using well-known and routine assays, such as those described in Section 5.11, infra.
In a preferred method, amino acid substitutions are introduced at one or more residues in the IgG constant domain or FcRn-binding fragment thereof and the mutated constant domains or fragments are expressed on the surface of bacteriophage which are then screened for increased FcRn binding affinity (see, in particular, Section 5.2 and 5.11, infra).

Preferably, the amino acid residues to be modified are surface exposed residues. Additionally, in making amino acid substitutions, preferably the amino acid residue to be substituted is a conservative amino acid substitution, for example, a polar residue is substituted with a polar residue, a hydrophilic residue with a hydrophilic residue, hydrophobic residue with a hydrophobic residue, a positively charged residue with a positively charged residue, or a negatively charged residue with a negatively charged residue. Moreover, preferably, the amino acid residue to be modified is not highly or completely conserved across species and/or is critical to maintain the constant domain tertiary structure or to FcRn binding. For example, but not by way of limitation, modification of the histidine at residue 310 is not preferred.
Specific mutants of the Fe domain that have increased affinity for FcRn were isolated after the third-round panning (as described in Section 6) from a library of mutant human IgGi molecules having mutations at residues 308-314 (histidine at position 310 and tryptophan at position 313 are fixed), those isolated after the fifth-round panning of the library for residues 251-256 (isoleucine at position 253 is fixed), those isolated after fourth-round panning of the library for residues 428-436 (histidine at position 429, glutamic acid at position 430, alanine at position 431, leucine at position 432, and histidine at position 435 are fixed), and those isolated after sixth-round panning of the library for residues 385-389 (glutamic acid at position 388 is fixed) are listed in Table I. The wild type human IgGl has a sequence Val-Leu-His-Gln-Asp-Trp-Leu (SEQ ID NO:86) at positions 308-314, Leu-Met-Ile-Ser-Arg-Thr (SEQ ID NO:87) at positions 251-256, Met-His-Glu-Ala-Leu-His-Asn-His-Tyr (SEQ ID NO:88) at positions 428-436, and Gly-Gln-Pro-Glu-Asn (SEQ ID
NO:89) at positions 385-389.
Table I
MUTANTS ISOLATED BY PANNING
LIBRARY 1 MUTANTS*

251-256 Leu Tyr Ile Thr Arg Glu (SEQ ID NO:90) Leu Tyr Ile Ser Arg Thr (SEQ ID NO:91) Leu Tyr Ile Ser Arg Ser (SEQ ID NO:92) Leu Tyr Ile SerArgArg (SEQ ID NO:93) Leu Tyr Ile Ser Arg Gin (SEQ ID NO:94) Leu Trp Ile Ser Arg Thr (SEQ ID NO:95) Leu Tyr Ile Ser Leu Gln (SEQ ID NO:96) Leu Phe Ile Ser Arg Asp (SEQ ID NO:97) Leu Phe Ile Ser Arg Thr (SEQ ID NO:98) Leu Phe Ile Ser Arg Arg (SEQ ID NO:99) Leu Phe Ile Thr Gly Ala (SEQ ID NO:100) Leu Ser Ile Ser Arg Glu (SEQ ID NO:101) Arg Thr Ile Ser Ile Ser (SEQ ID NO:102) 308-314 Thr Pro His Ser Asp Trp Leu (SEQ ID NO:103) Ile Pro His Glu Asp Trp Leu (SEQ ID NO:104) 385-389 Arg Thr Arg Glu Pro (SEQ ID NO: 105) Asp Pro Pro Glu Ser (SEQ ID NO:106) Ser Asp Pro Glu Pro (SEQ ID NO:107) Thr Ser His Glu Asn (SEQ ID NO:108) Ser Lys Ser Glu Asn (SEQ ID NO:109) His Arg Ser Glu Asn (SEQ ID NO: 110) Lys Ile Arg Glu Asn (SEQ ID NO:111) LIBRARY MUTANTS*

Gly Ile Thr Glu Ser (SEQ ID NO: 112) Ser Met Ala Glu Pro (SEQ ID NO:113) 428-436 Met His Glu Ala LeuArg Tyr His His (SEQ ID NO: 114) Met His Glu Ala Leu His Phe His His (SEQ ID NO: 115) Met His Glu Ala Leu Lys Phe His His (SEQ ID NO:116) Met His Glu Ala Leu Ser Tyr His Arg (SEQ ID NO: 117) Thr His Glu Ala Leu His Tyr His Thr (SEQ ID NO: 118) Met His Glu Ala Leu His Tyr His Tyr (SEQ ID NO:119) * Substituting residues are indicated in bold face The underlined sequences in Table I correspond to sequences that occurred 10 to 20 times in the final round of panning and the sequences in italics correspond to sequences that occurred 2 to 5 times in the final round of panning. Those sequences that are neither underlined nor italicized occurred once in the final round of panning.
In one preferred embodiment, the invention provides modified immunoglobulin molecules (e.g., various antibodies) that have increased in vivo half-life and affinity for FcRn relative to unmodified molecules (and, in preferred embodiments, altered bioavailabilty such as increased or decreased transport to mucosal surfaces or other target tissues). Such immunoglobulin molecules include IgG molecules that naturally contain an FcRn binding domain and other non-IgG immunoglobulins (e.g., IgE, IgM, IgD, IgA and IgY) or fragments of immunoglobulins that have been engineered to contain an FeRn-binding fragment (i.e., fusion proteins comprising non-IgG immunoglobulin or a portion thereof and an FcRn binding domain). In both cases the FcRn-binding domain has one or more amino acid modifications that increase the affinity of the constant domain fragment for FcRn.
The modified immunoglobulins include any immunoglobulin molecule that binds (preferably, immunospecifically, i.e., competes off non-specific binding), as determined by immunoassays well known in the art for assaying specific antigen-antibody binding) an antigen and contains an FcRn-binding fragment. Such antibodies include, but are not limited to, polyclonal, monoclonal, bi-specific, multi-specific, human, humanized, chimeric antibodies, single chain antibodies, Fab fragments, F(ab')2 fragments, disulfide-linked Fvs, and fragments containing either a VL or VH domain or even a complementary determining region (CDR) that specifically binds an antigen, in certain cases, engineered to contain or fused to an FcRn binding domain.
The IgG molecules of the invention, and FcRn-binding fragments thereof, are preferably IgGi subclass of IgGs, but may also be any other IgG subclasses of given animals. For example, in humans, the IgG class includes IgGl, IgG2, IgG3, and IgG4; and mouse IgG includes IgGl, IgG2a, IgG2b, IgG2c and IgG3. It is known that certain IgG
subclasses, for example, mouse IgG2b and IgG2c, have higher clearance rates than, for example, IgGl (Medesan et al., Eur. J Immunol., 28:2092-2100, 1998). Thus, when using IgG subclasses other than IgG1, it may be advantageous to substitute one or more of the residues, particularly in the CH2 and CH3 domains, that differ from the IgG1 sequence with those of IgGl, thereby increasing the in vivo half-life of the other types of IgG.
The immunoglobulins (and other proteins used herein) may be from any animal origin including birds and mammals. Preferably, the antibodies are human, rodent (e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken. As used herein, "human" antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins, as described infra and, for example, in U.S. Patent No. 5,939,598 by Kucherlapati et al.
The antibodies of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopes of a polypeptide or may be specific for heterologous epitopes, such as a heterologous polypeptide or solid support material. See, e.g., PCT
publications WO
93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J Immunol., 147:60-69, 1991; U.S. Patent Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920;
5,601,819;
Kostelny et al., I Immunol., 148:1547-1553, 1992.
The antibodies of the invention include derivatives that are otherwise modified, i.e., by the covalent attachment of any type of molecule to the antibody such that covalent attachment does not prevent the antibody from binding antigen and/or generating an anti-idiotypic response. For example, but not by way of limitation, the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications maybe carried out by known techniques, including, but not limited to, specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non classical amino acids.
Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. For example, monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas, pp. 563-681 (Elsevier, N.Y., 1981). The term "monoclonal antibody"
as used herein is not limited to antibodies produced through hybridoma technology. The term "monoclonal antibody" refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.

Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art. In a non-limiting example, mice can be immunized with an antigen of interest or a cell expressing such an antigen.
Once an immune response is detected, e.g., antibodies specific for the antigen are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated.
The splenocytes are then fused by well known techniques to any suitable myeloma cells.
Hybridomas are selected and cloned by limiting dilution. The hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding the antigen. Ascites fluid, which generally contains high levels of antibodies, can be generated by inoculating mice intraperitoneally with positive hybridoma clones., Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, Fab and F(ab')2 fragments may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). F(ab')2 fragments contain the complete light chain, and the variable region, the CH1 region and the hinge region of the heavy chain.
For example, antibodies can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. In a particular embodiment, such phage can be utilized to display antigen binding domains, such as Fab and Fv or disulfide-bond stabilized Fv, expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
Phage used in these methods are typically filamentous phage, including fd and M13. The antigen binding domains are expressed as a recombinantly fused protein to either the phage gene III or gene VIII protein. Alternatively, the modified FcRn binding portion of immunoglobulins of the present invention can be also expressed in a phage display system.
Examples of phage display methods that can be used to make the immunoglobulins, or fragments thereof, of the present invention include those disclosed in Brinkman et al., J.
Immunol. Methods, 182:41-50, 1995; Ames et al., J. Immunol. Methods, 184:177-186, 1995; Kettleborough et al., Eur. J. Immunol., 24:952-958, 1994; Persic et al., Gene, 187:9-18, 1997; Burton et al., Advances in Immunology, 57:191-280, 1994; PCT
publications WO 90/02809; WO 91110737; WO 92/01047; WO 92/18619; WO 93/11236; WO 95/15982;
WO 95/20401; and U.S. Patent Nos. 5,698,426; 5,223,409; 5,403,484; 5,580,717;
5,427,908;
5,750,753; 5,821,047; 5,571,698; 5,427,908; 5,516,637; 5,780,225; 5,658,727;
5,733,743 and 5,969,108.
As described in the above references, after phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired fragments, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below. For example, techniques to recombinantly produce Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et al., BioTechniques, 12(6):864-869, 1992; and Sawai et al., AJRI, 34:26-34, 1995; and Better et al., Science, 240:1041-1043, 1988. Examples of techniques which can be used to produce single-chain Fvs and antibodies include those described in U.S. Patent Nos. 4,946,778 and 5,258,498; Huston et al., Methods in Enzymology, 203:46-88, 1991; Shu et al., PNAS, 90:7995-7999, 1993; and Skerra et al., Science, 240:1038-1040, 1988.
For some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use chimeric, humanized, or human antibodies. A
chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a constant region derived from a human immunoglobulin.
Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science, 229:1202, 1985; Oi et al., BioTechniques, 4:214 1986; Gillies et al., J. Immunol.
Methods, 125:191-202, 1989; U.S. Patent Nos. 5,807,715; 4,816,567; and 4,816,397.
Humanized antibodies are antibody molecules from non-human species that bind the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule.
Often, framework residues in the human framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. See, e.g., Queen et al., U.S. Patent No.
5,585,089; Riechmann et al., Nature, 332:323, 1988. Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400;
PCT publication WO 91/09967; U.S. Patent Nos. 5,225,539; 5,530,101 and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology, 28(4/5):489-498, 1991; Studnicka et al., Protein Engineering, 7(6):805-814, 1994;
Roguska et al., Proc Natl. Acad. Sci. USA, 91:969-973, 1994), and chain shuffling (U.S.
Patent No. 5,565,332).
Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See U.S. Patent Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645; WO 98/50433; WO 98/24893; WO
98/16654; WO 96/34096; WO 96/33735; and WO 91/10741.
Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. For an overview of this technology for producing human antibodies, see Lonberg and Huszar, Int. Rev. Immunol., 13:65-93, 1995. For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., PCT
publications WO
98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0 598 877;
U.S. Patent Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825; 5,661,016; 5,545 806;

5,814,318; 5,885,793; 5,916,771; and 5,939,598. In addition, companies such as Abgenix, Inc. (Fremont, CA), Medarex (NJ) and Genpharm (San Jose, CA) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.
Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection." In this approach a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope. (Jespers et al., Bioltechnology, 12:899-903, 1988).
In particular embodiments, the modified antibodies have in vivo therapeutic and/or prophylactic uses. Examples of therapeutic and prophylactic antibodies which may be so modified include, but are not limited to, SYNAGIS (Medlmmune, MD) which is a humanized anti-respiratory syncytial virus (RSV) monoclonal antibody for the treatment of patients with RSV infection; HERCEPTIN (Trastuzumab) (Genentech, CA) which is a humanized anti-HER2 monoclonal antibody for the treatment of patients with metastatic breast cancer; REMICADE (infliximab) (Centocor, PA) which is a chimeric anti-TNFa monoclonal antibody for the treatment of patients with Crone's disease; REOPRO

(abciximab) (Centocor) which is an anti-glycoprotein IIb/lIIa receptor on the platelets for the prevention of clot formation; ZENAPAX (daclizumab) (Roche Pharmaceuticals, Switzerland) which is an immunosuppressive, humanized anti-CD25 monoclonal antibody for the prevention of acute renal allograft rejection. Other examples are a humanized anti-CD 18 F(ab')2 (Genentech); CDP860 which is a humanized anti-CD 18 F(ab')2 (Celltech, UK); PRO542 which is an anti-HIV gp120 antibody fused with CD4 (Progenics/Genzyme Transgenics); Ostavir which is a human anti Hepatitis B virus antibody (Protein Design Lab/Novartis); PROTOVIRTM which is a humanized anti-CMV IgGi antibody (Protein Design Lab/Novartis); MAK-195 (SEGARD) which is a murine anti-TNF-a F(ab')2 (Knoll Pharma/BASF); IC14 which is an anti-CD14 antibody (ICOS Pharm); a humanized anti-VEGF IgGi antibody (Genentech); OVAREXTM which is a murine anti-CA 125 antibody (Altarex); PANOREXTM which is a murine anti-17-IA cell surface antigen IgG2a antibody (Glaxo Wellcome/Centocor); BEC2 which is a murine anti-idiotype (GD3 epitope) IgG
antibody (ImClone System); IMC-0225 which is a chimeric anti-EGFR IgG antibody (ImClone System); VITAXINTM which is a humanized anti-aVf33 integrin antibody (Applied Molecular Evolution/Medimmune); Campath IH/LDP-03 which is a humanized anti CD52 IgG1 antibody (Leukosite); Smart M195 which is a humanized anti-CD33 IgG
antibody (Protein Design Lab/Kanebo); RITUXANTM which is a chimeric anti-CD20 IgG1 antibody (IDEC Pharm/Genentech, Roche/Zettyaku); LYMPHOCIDETM which is a humanized anti-CD22 IgG antibody (Immunomedics); Smart ID 10 which is a humanized anti-HLA antibody (Protein Design Lab); ONCOLYMTM (Lym-1) is a radiolabelled murine anti-HLA DIAGNOSTIC REAGENT antibody (Techniclone); ABX-IL8 is a human anti-IL8 antibody (Abgenix); anti-CD 11 a is a humanized IgG1 antibody (Genetech/Xoma);
ICM3 is a humanized anti-ICAM3 antibody (ICOS Pharm); IDEC-1 14 is a primatied anti-CD80 antibody (IDEC Pharm/Mitsubishi); ZEVALINTM is a radiolabelled murine anti-CD20 antibody (IDEC/Schering AG); IDEC-131 is a humanized anti-CD40L antibody (IDEC/Eisai); IDEC-151 is a primatized anti-CD4 antibody (IDEC); IDEC-152 is a primatized anti-CD23 antibody (IDEC/Seikagaku); SMART anti-CD3 is a humanized anti-CD3 IgG (Protein Design Lab); 5G1.1 is a humanized anti-complement factor 5 (C5) antibody (Alexion Pharm); D2E7 is a humanized anti-TNF-a antibody (CAT/BASF);
CDP870 is a humanized anti-TNF-a Fab fragment (Celltech); IDEC- 151 is a primatized anti-CD4 IgG1 antibody (IDEC Pharm/SmithKline Beecham); MDX-CD4 is a human anti-CD4 IgG antibody (Medarex/Eisai/Genmab); CDP571 is a humanized anti-TNF-a IgG4 antibody (Celltech); LDP-02 is a humanized anti-a407 antibody (LeukoSite/Genentech);
OrthoClone OKT4A is a humanized anti-CD4 IgG antibody (Ortho Biotech);
ANTOVATM
is a humanized anti-CD40L IgG antibody (Biogen); ANTEGRENTM is a humanized anti-VLA-4 IgG antibody (Elan); MDX-33 is a human anti-CD64 (FcyR) antibody (Medarex/Centeon); SCH55700 is a humanized anti-IL-S IgG4 antibody (Celltech/Schering); SB-240563 and SB-240683 are humanized anti-IL-5 and IL-4 antibodies, respectively, (SmithKline Beecham); rhuMab-E25 is a humanized anti-IgE IgGI
antibody (Genentech/Norvartis/Tanox Biosystems); IDEC-152 is a primatized anti-antibody (IDEC Pharm); ABX-CBL is a murine anti CD-147 IgM antibody (Abgenix);
BTI-322 is a rat anti-CD2 IgG antibody (Medimmune/Bio Transplant); Orthoclone/OKT3 is a murine anti-CD3 IgG2a antibody (ortho Biotech); SIMULECTTM is a chimeric anti-IgG1 antibody (Novartis Pharm); LDP-01 is a humanized anti-J32-integrin IgG
antibody (Leuko Site); Anti-LFA- 1 is a murine anti CD 18 F(ab')2 (Pasteur-Merieux/hnmunotech);
CAT-152 is a human anti-TGF-(32 antibody (Cambridge Ab Tech); and Corsevin M
is a chimeric anti-Factor VII antibody (Centocor).
In specific embodiments, the invention provides modified antibodies having one or more of the mutations described herein and that immunospecifically bind RSV, e.g., SYNAGIS . The present invention also provides modified antibodies having one or more of the mutations described herein and that comprise a variable heavy (VH) and/or variable light (VL) domain having the amino acid sequence of any VH and/or VL domain listed in Table III. The present invention further encompasses anti-RSV antibodies comprising one or more VH complementarity determining regions (CDRs) and/or one or more VL
CDRs having the amino acid sequence of one or more VH CDRs and/or VL CDRS listed in Table III or one or more of the CDRs listed in Table II wherein one or more of the bolded and underlined residues has an amino acid substitution, preferably that increases the affinity of the antibody for RSV. In specific embodiments, the antibody to be modified is AFFF, pl2f2, p12f4, plld4, A1e109, A12a6, A13c4, A17d4, A4B4, A8C7, 1X-493L1FR, H3-3F4, M3H9, Y10H6, DG, AFFF(l), 6H8, L1-7E5, L215B10, A13A11, A1H5, A4B4(l), A4B4L1FR-S28R, A4B4-F52S.
Table II.
CDR Sequences of SYNAGIS
CDR Sequence SEQ ID NO:

c o w , c h F, c F w H o F
rF` O w 0 0 Fw O w O w O w O w o ~` O
ca ~z z z >z ~z z >z >z z U Q Q (~ Q Q
0 a 0 a 0 a a a 0 a O a 0 a a w d a a w w a w a w a w d w a w w n 4r Z w v V. a. w v~ w v w '07i 0 z z ac# 1 z al z Z va~l z vaa~l z z AI R FI R III Q FI (~ HI FI Q Q
Q o Q o a a Q a a a Q a Q a d - N N N N
~ O N N ~ M N M N d:
0Z OZ 0Z ;Z C7Z 0Z O^Z OZ O Zn el-w m rn v2 v~ vs d la la la I
a Ia la la la G ti N N C> M M dN', V
o z z z z Z z z z z a a a a a a a a a cn r~ M Q m A '" o N Q N Q N Q N Q N Q N Q N
a Z wiz wiz wiz z 1z w1z lz E x~ a & pia wia ,a is wia pia la F 12 a AI E AI s Al s AI .mow. AI s AI Es A- Fw En &D ~~ In cl, cl, N N N N N M V fit;
A z z Iz IZ Iz Iz O Z ziZ ziz Q a 0l a Ol a Ol a cal a cal a Q a Q Q A Q 0 A Q A fQ

0 71 ,^ o 00 0~`~0 ' 'n 0 0 0 q Z Z c z C Z Z Z CO Z Z on z o ~0I a aI a aI a I a a dI a a a H h W H v H w w F w Es Es F w F ri '~^1 R n O~ F7 N N R M R rn ^~ r+ v'ct.
E a0 ao a0 oao a0 0'0 0' 0'0 ad p z rwiD Z vwi Z Z vwi Z z vwi z ~' C7 w ~r o 0 v `to N
Z ¾~ a a o E . .o F. ~o H ro f~ ~o I- ~o w a w F ~o F 0 E- o a O w O ts. O a O u' O t, O O O O 11-0 IL. O O
z z z z z z z ~z z z c7 t7 t7 o c7 c7 0 wl cti c7 o c7 ~~ W~
0a d/a na av oa c~ c~~ya 0 a c~a c~a a aw Ow aw aw aw aw Ow aw c'w aw aw aw wv w wv wti, wv wv wv ww u.) wQ wv wv O c=1 N ~
N N y N N -'+ t0 W O
q~ Qt z o Z Z 2 z Z Z z z Z Z
UQ C41Q QIQ~Q IQIQfQla v.18 Q
Auaj A Aw Aw Aw QN Aw Aw Aw Qw Aw Aw A Z Oz O z C7z Z Oz O z O7... 02 Oz ( z OZ

a g a go, a a ~, a a a a ~~ww ~~w "' ww w .w w w ¾ ww w w f~ii~)II v v VJ v v v VJ v v1 v VJ v id v VJ v et QN .1 W N co 'O O N '' N co 1õ' !h N N N .~ N N ~6 O D ~O O

o z z z z z z z z z z z z a 8 ~ Q 8 0 ~ ~ ~ Q 8 8 8 w w w w O' w w w =w w w Cr v~ v~ ~n w rn h ,.v) Cl) > o > o. > rn > > > rn > > > rn > >
a N A N A A A N (~ [~ A p r (a A n A N
0 0. a w, zz zz z Z Z Z z z , zo IW Z k,, Z Z
I0 8 ER 2 G I8 Q 8 8 a 8 A Q
SCI a o' a a a a a a a a a S AS is AI w~, v~l al QI s Ai~w. v, .a a -a a ..a ..a a Qj .a ..a 0) rn Go V) V3 tz aisz z z z z z z z z z z 2 Q 2Q 2P 212 2G Rs Re 92 e Aw r Aw Aw PAw BY (~A~^ W Aw A~^w PAw Aw PAw q A Q A A A A A Q
a o ^r o 0 C7 0 0 0^ 0 0 0 '6 O .. CJ C7 r, C7 'O" ;7 > o > C7 .. c7 ..
a >o > > >o >o >o >O > >O >o >o Z ~Z yz CZ ~Z C02 Z Nz f Z

~q ~8q ae oQ o~ oa ~q eta eta y o' eta eta et eta et a et a eta eta eta a H E- w H w ~+ w 1- w t- w t- w H w E- w N w E- w 03 rn v f/! So H v' h VJ tt2 m 3 v v ^^ (n~ v (~ v ~ v [~ v v (~ v nn v nn v w 8 h G h G y = h 8 'CI G7 F7 O) CI h F7 A t7 b o W p p o z O W p O w p o' O o' p a p a A qz Wz yz 07z z Z z Gz wz yz v ^ N o 00 M x .o c Ch w x Q
z a a y A ~, H ? H ? H
A zz zz zz z coo as a as as Al z z Al z ~z A wl ~ A o~ Q a Q ~' Q a ti ~ > ~ a ~ a Q a ~
a a co a a e z z 0 z A f? Q
a a a a M O N Q N Q N Q N
a a a a AI~% AID AID AI`/
~I I I ~I
a a a a a Iz Izz :iz Iz Ia Aa Qa caa Q ra Q Q

0 7~ 71 (D 71 0 7i ~z z ~z 0 ~z a ~I
a a dl a 00 2 cc A 0 0 a 0 0 00 co In other embodiments, the antibody is a modified anti-a(33 antibody, preferably a Vitaxin antibody (see, PCT publications WO 98/33919 and WO
00/78815, both by Huse et al..
Modified IgGs of the present invention having longer half-lives than wild type may also include IgGs whose bioactive sites, such as antigen-binding sites, Fe-receptor binding sites, or complement-binding sites, are modified by genetic engineering to increase or reduce such activities compared to the wild type.
Modification of these and other therapeutic antibodies to increase the in vivo half-life permits administration of lower effective dosages and/or less frequent dosing of the therapeutic antibody. Such modification to increase in vivo half-life can also be useful to improve diagnostic immunoglobulins as well, for example, permitting administration of lower doses to achieve sufficient diagnostic sensitivity.
The present invention also provides fusion proteins comprising a bioactive molecule and an hinge-Fc region or a fragment thereof (preferably human) having one or more modifications (Le., substitutions, deletions, or insertions)in amino acid residues identified to be involved in the interaction between the hinge-Fc region and the FcRn receptor. In particular, the present invention provides fusion proteins comprising a bioactive molecule recombinantly fused or chemically conjugated (including both covalent and non-covalent conjugations) to a CH2 domain having one or more modifications in amino acid residues 251-256, 285-290, and/or amino acid residues 308-314, and/or to a CH3 domain having one or more modifications in amino acid residues 385-389 and/or 428-436, in particular, one or more of the amino acid substitutions discussed above. The fusion of a bioactive molecule to a constant domain or a fragment thereof with one or more of such modifications increases the in vivo half-life of the bioactive molecule.
In a preferred embodiment, fusion proteins of the invention comprise a bioactive molecule recombinantly fused or chemically conjugated to a CH2 domain having one or more amino acid residue substitutions in amino acid residues 251-256, 285-290, and/or amino acid residues 308-314, and/or to a CH3 domain having one or more modifications in amino acid residues 385-389 and/or 428-436. In certain embodiments, a fusion protein comprises a CH2 domain of IgG molecule in which amino acid residues 253, 310, and 313 are not modified. In another embodiment, a fusion protein comprises a CH3 domain of IgG molecule in which Amino acid residues 388, 429, 430, 431, 432, and 435 are not modified.
A bioactive molecule can be any polypeptide or synthetic drug known to one of skill in the art. Preferably, a bioactive molecule is a polypeptide consisting of at least 5, preferably at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acid residues. Examples of bioactive polypeptides include, but are not limited to, various types of antibodies, cytokines (e.g., IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-10, IL-12, IL-15, IFN-y, IFN-a, and IFN-(3), cell adhesion molecules (e.g., CTLA4, CD2, and CD28), ligands (e.g., TNF-a,, TNF-(3, and an anti-angiogenic factor such as endostatin), receptors, antibodies and growth factors (e.g., PDGF, EGF, NGF, and KGF).
A bioactive molecule can also be a therapeutic moiety such as a cytotoxin (e.g., a cytostatic or cytocidal agent), a therapeutic agent or a radioactive element (e.g, alpha-emitters, gamma-emitters, etc.). Examples of cytostatic or cytocidal agents include, but are not limited to, paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cisdichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
The present invention also provides polynucleotides comprising a nucleotide sequence encoding a modified IgG of the invention and fragments thereof which contain the modified FcRn binding sites with increased affinity and vectors comprising said polynucleotides. Furthermore, the invention includes polynucleotides that hybridize under stringent or lower stringent hybridization conditions to polynucleotides encoding modified IgGs of the present invention.
The nucleotide sequence of modified IgGs and the polynucleotides encoding the same may be obtained by any methods known in the art, including general DNA
sequencing method, such as dideoxy chain termination method (Sanger sequencing), and oligonucleotide priming in combination with PCR, respectively.

5.2. IDENTIFICATION OF MUTATIONS WITHIN
THE HINGE-FC REGION OF IMMUNOGLOBULIN
MOLECULES

One or more modifications in amino acid residues 251-256, 285-290, 308-314, 385-389, and 428-436 of the constant domain may be introduced utilizing any technique known to those of skill in the art. The constant domain or fragment thereof having one or more modifications in amino acid residues 251-256, 285-290, 308-314, 385-389, and 428-436 may be screened by, for example, a binding assay to identify the constant domain or fragment thereof with increased affinity for the FcRn receptor (e.g., as described in section 5.11, infra). Those modifications in the hinge-Fe domain or the fragments thereof which increase the affinity of the constant domain or fragment thereof for the FcRn receptor can be introduced into antibodies to increase the in vivo half-lives of said antibodies. Further, those modifications in the constant domain or the fragment thereof which increase the affinity of the constant domain or fragment thereof for the FcRn can be fused to bioactive molecules to increase the in vivo half-lives of said bioactive molecules (and, preferably alter (increase or decrease) the bioavailability of the molecule, for example, to increase or decrease transport to mucosal surfaces (or other target tissue) (e.g., the lungs)).

5.2.1. MUTAGENESIS
Mutagenesis may be performed in accordance with any of the techniques known in the art including, but not limited to, synthesizing an oligonucleotide having one or more modifications within the sequence of the constant domain of an antibody or a fragment thereof (e.g., the CH2 or CH3 domain) to be modified. Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed.
Typically, a primer of about 17 to about 75 nucleotides or more in length is preferred, with about 10 to about 25 or more residues on both sides of the junction of the sequence being altered. A number of such primers introducing a variety of different mutations at one or more positions may be used to generate a library of mutants.
The technique of site-specific mutagenesis is well known in the art, as exemplified by various publications (see, e.g., Kunkel et al., Methods Enzymol., 154:367-82, 1987). In general, site-directed mutagenesis is performed by first obtaining a single-stranded vector or melting apart of two strands of a double stranded vector which includes within its sequence a DNA
sequence which encodes the desired peptide. An oligonucleotide primer bearing the desired mutated sequence is prepared, generally synthetically. This primer is then annealed with the single-stranded vector, and subjected to DNA polymerizing enzymes such as T7 DNA
polymerase, in order to complete the synthesis of the mutation-bearing strand.
Thus, a heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform or transfect appropriate cells, such as E. coil cells, and clones are selected which include recombinant vectors bearing the mutated sequence arrangement. As will be appreciated, the technique typically employs a phage vector which exists in both a single stranded and double stranded form. Typical vectors useful in site-directed mutagenesis include vectors such as the M13 phage. These phage are readily commercially available and their use is generally well known to those skilled in the art. Double stranded plasmids are also routinely employed in site directed mutagenesis which eliminates the step of transferring the gene of interest from a plasmid to a phage.
Alternatively, the use of PCRTM with commercially available thermostable enzymes such as Taq DNA polymerase may be used to incorporate a mutagenic oligonucleotide primer into an amplified DNA fragment that can then be cloned into an appropriate cloning or expression vector. See, e.g., Tomic et al., Nucleic Acids Res., 18(6):1656, 1987, and Upender et al., Biotechniques, 18(1):29-30, 32, 1995, for PCRTM -mediated mutagenesis procedures. PCRTM employing a thermostable ligase in addition to a thermostable polymerase may also be used to incorporate a phosphorylated mutagenic oligonucleotide into an amplified DNA fragment that may then be cloned into an appropriate cloning or expression vector (see e.g., Michael, Biotechniques, 16(3):410-2, 1994).
Other methods known to those of skill in the art of producing sequence variants of the Fc domain of an antibody or a fragment thereof can be used.
For example, recombinant vectors encoding the amino acid sequence of the constant domain of an antibody or a fragment thereof may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants.
5.2.2. PANNING
Vectors, in particular, phage, expressing constant domains or fragments thereof having one or more modifications in amino acid residues 251-256,285-290,308-314, 385-389, and/or 428-436 can be screened to identify constant domains or fragments thereof having increased affinity for FcRn to select out the highest affinity binders from a population of phage. Immunoassays which can be used to analyze binding of the constant domain or fragment thereof having one or more modifications in amino acid residues 251-256, 285-290, 308-314, 385-389, and/or 428-436 to the FcRn include, but are not limited to, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich"
immunoassays, and fluorescent immunoassays. Such assays are routine and well known in the art (see, e.g., Ausubel et al., eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York). Exemplary immunoassays are described briefly herein below (but are not intended by way of limitation). BlAcore kinetic analysis can also be used to determine the binding on and off rates of a constant domain or a fragment thereof having one or more modifications in amino acid residues 251-256, 285-290, 308-314, 385-389, and/or 428-436 to the FeRn. BlAcore kinetic analysis comprises analyzing the binding and dissociation of a constant domain or a fragment thereof having one or more modifications in amino acid residues 251-256, 285-290,308-314,385-389, and/or 428-436 from chips with immobilized FcRn on their surface (see section 5.1 and the Example section infra).

5.2.3. SEQUENCING
Any of a variety of sequencing reactions known in the art can be used to directly sequence the nucleotide sequence encoding constant domains or fragments thereof having one or more modifications in amino acid residues 251-256, 285-290, 308-314, 385-389, and/or 428-436. Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert (Proc. Natl. Acad. Sci. USA, 74:560, 1977) or Sanger (Proc. Natl. Acad Sci. USA, 74:5463, 1977). It is also contemplated that any of a variety of automated sequencing procedures can be utilized (Bio/Techniques, 19:448, 1995), including sequencing by mass spectrometry (see, e.g., PCT Publication No. WO 94/16101, Cohen et al., Adv. Chromatogr., 36:127-162, 1996, and Griffin et al., Appl. Biochem.
Biotechnol., 38:147-159, 1993).

5.3. RECOMBINANT METHODS OF PRODUCING
ANTIBODIES

The antibodies of the invention or fragments thereof can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques.

The nucleotide sequence encoding an antibody may be obtained from any information available to those of skill in the art (i. e., from Genbank, the literature, or by routine cloning). If a clone containing a nucleic acid encoding a particular antibody or an epitope-binding fragment thereof is not available, but the sequence of the antibody molecule or epitope-binding fragment thereof is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA
library, or a cDNA library generated from, or nucleic acid, preferably poly AA
RNA, isolated from any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers hybridizable to the 3' and 5 'ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody.
Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art.
Once the nucleotide sequence of the antibody is determined, the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY; and Ausubel et al., eds., 1998, Current Protocols in Molecular Biology, John Wiley & Sons, NY), to generate antibodies having a different amino acid sequence by, for example, introducing amino acid substitutions, deletions, and/or insertions into the epitope-binding domain regions of the antibodies and preferably, into the hinge-Fc regions of the antibodies which are involved in the interaction with the FcRn. In a preferred embodiment, antibodies having one or more modifications in amino acid residues 251-256, 285-290, 308-314, 385-389, and 428-436 are generated.

Recombinant expression of an antibody requires construction of an expression vector containing a nucleotide sequence that encodes the antibody. Once a nucleotide sequence encoding an antibody molecule or a heavy or light chain of an antibody, or portion thereof (preferably, but not necessarily, containing the heavy or light chain variable region) has been obtained, the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, provides replicable vectors comprising a nucleotide sequence encoding the constant region of the antibody molecule with one or more modifications in the amino acid residues involved in the interaction with the FcRn (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Patent No.
5,122,464). The nucleotide sequence encoding the heavy-chain variable region, light-chain variable region, both the heavy-chain and light-chain variable regions, an epitope-binding fragment of the heavy- and/or light-chain variable region, or one or more complementarity determining regions (CDRs) of an antibody may be cloned into such a vector for expression.
The expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody having an increased affinity for the FcRn and an increased in vivo half-life.
Thus, the invention includes host cells containing a polynucleotide encoding an antibody, a constant domain or a FcRn binding fragment thereof having one or more modifications in amino acid residues 251-256, 285-290, 308-314, 385-389, and/or 428-436, preferably, operably linked to a heterologous promoter.
A variety of host-expression vector systems may be utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ. These include, but are not limited to, microorganisms such as bacteria (e.g., E. coli and B.
subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces and Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; and tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; and mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 and NSO cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K
promoter). Preferably, bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene, 45:101, 1986, and Cockett et al., Bio/Technology, 8:2, 1990).
In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody molecule, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited to, the E. coli expression vector pUR278 (Ruther et al., EMBO, 12:1791, 1983), in which the antibody coding sequence may be ligated individually into the vector in frame with the lacZ coding region so that a fusion protein is produced; and pIN vectors (Inouye & Inouye, Nucleic Acids Res., 13:3101-3109, 1985 and Van Heeke &
Schuster, J. Biol. Chem., 24:5503-5509, 1989).
In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera fi=ugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
In mammalian host cells, a number of viral-based expression systems may be utilized to express an antibody molecule of the invention. In cases where an adenovirus is used as an expression vector, the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts (e.g., see Logan & Shenk, Proc. Natl.
Acad. Sci. USA, 81:355-359, 1984). Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG
initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc.
(see, e.g., Bitter et al., Methods in Enzymol., 153:516-544, 1987).
In addition, a host cell strain may be chosen which modulates the expression of the antibody sequences, or modifies and processes the antibody in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the antibody. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the antibody expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include but are not limited to CHO, VERY, BHK, HeLa, COS, MDCK, 293, 3T3, W 138, and in particular, myeloma cells such as NSO cells, and related cell lines, see, for example, Morrison et al., U.S. Patent No. 5,807,715.

For long-term, high-yield production of recombinant antibodies, stable expression is preferred. For example, cell lines which stably express the antibody molecule may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid - into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the antibody molecule. Such engineered cell lines may be particularly useful in screening and evaluation of compositions that interact directly or indirectly with the antibody molecule.
A number of selection systems may be used, including but not limited to, the herpes simplex virus thymidine kinase (Wigler et al., Cell; 11:223, 1977), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA, 48:202, 1992), and adenine phosphoribosyltransferase (Lowy et al., Cell, 22:8-17, 1980) genes can be employed in tk, hgprt or aprt cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following; genes:
dhfr, which confers resistance to methotrexate (Wigler et al., Natl. Acad. Sci. USA, 77:357. 1980 and O'Hare et al., Proc. Natl. Acad. Sci. USA, 78:1527, 1981); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA, 78:2072, 1981); neo, which confers resistance to the aminoglycoside G-418 (Wu and Wu, Biotherapy, 3:87-95, 1991; Tolstoshev, Ann. Rev. Pharmacol. Toxicol., 32:573-596, 1993; Mulligan, Science, 260:926-932, 1993; and Morgan and Anderson, Ann. Rev. Biochem., 62: 191-217, 1993; and May, TIB TECH, 11(5):155-2 15, 1993); and hygro, which confers resistance to hygromycin (Santerre et al., Gene, 30:147, 1984). Methods commonly known in the art of recombinant DNA technology may be routinely applied to select the desired recombinant clone, and such methods are described, for example, in Ausubel et al. (eds.), 1993, Current Protocols in Molecular Biology, John Wiley & Sons, NY; Kriegler, 1990, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY; in Chapters 12 and 13, Dracopoli et al. (eds), 1994, Current Protocols in Human Genetics, John Wiley & Sons, NY; and Colberre-Garapin et al., J. Mol. Biol., 150:1, 1981.
The expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, 1987, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. Academic Press, New York). When a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., Mol., Cell. Biol., 3:257,1983).
The host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides or different selectable markers to ensure maintenance of both plasmids. Alternatively, a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature, 322:52, 1986;
and Kohler, Proc. Natl. Acad. Sci. USA, 77:2 197, 1980). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
Once an antibody molecule of the invention has been produced by recombinant expression, it may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A
purification, and sizing column chromatography), centrifugation, differential solubility, or by any other standard techniques for the purification of proteins. Further, the antibodies of the present invention or fragments thereof may be fused to heterologous polypeptide sequences described herein or otherwise known in the art to facilitate purification.

5.3.1. ANTIBODY CONJUGATES
The present invention encompasses antibodies recombinantly fused or chemically conjugated (including both covalently and non-covalently conjugations) to heterologous polypeptides (i.e., an unrelated polypeptide; or portion thereof, preferably at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids of the polypeptide) to generate fusion proteins. The fusion does not necessarily need to be direct, but may occur through linker sequences.
Antibodies fused or conjugated to heterologous polypeptides may also be used in in vitro immunoassays and purification methods using methods known in the art. See e.g., PCT
Publication No. WO 93/21232; EP 439,095; Naramura et al., Immunol. Lett., 39:91-99, 1994; U.S. Patent 5,474,981; Gillies et al., PNAS, 89:1428-1432, 1992; and Fell et al., J.
Immunol., 146:2446-2452, 1991.

Antibodies can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci. USA, 86:821-824, 1989, for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell, 37:767 1984) and the "flag" tag (Knappik et al., Biotechniques, 17(4):754-761, 1994).
The present invention also encompasses antibodies conjugated to a diagnostic or therapeutic agent or any other molecule for which in vivo half-life is desired to be increased. The antibodies can be used diagnostically to, for example, monitor the development or progression of a disease, disorder or infection as part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen.
Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals, and nonradioactive paramagnetic metal ions. The detectable substance may be coupled or conjugated either directly to the antibody or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, 0-galactosidase, or acetylcholinesterase;
examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include 1251, 1311, "'In or '9'nTc.
An antibody may be conjugated to a therapeutic moiety such as a cytotoxin (e.g., a cytostatic or cytocidal agent), a therapeutic agent or a radioactive element (e.g., alpha-emitters, gamma-emitters, etc.). Cytotoxins or cytotoxic agents include any agent that is detrimental to cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, I -dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cisdichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
Further, an antibody may be conjugated to a therapeutic agent or drug moiety that modifies a given biological response. Therapeutic agents or drug moieties are not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, a-interferon (IFN-a), (3-interferon (IFN-(3), nerve growth factor (NGF), platelet derived growth factor (PDGF), tissue plasminogen activator (TPA), an apoptotic agent (e.g., TNF-a, TNF-3, AIM I as disclosed in PCT
Publication No. WO 97/33899), AIM II (see, PCT Publication No. WO 97/34911), Fas Ligand (Takahashi et al., J. Iminunol., 6:1567-1574, 1994), and VEGI (PCT
Publication No.
WO 99/23105), a thrombotic agent or an anti-angiogenic agent (e.g., angiostatin or endostatin); or a biological response modifier such as, for example, a lymphokine (e.g., interleukin-1 ("IL- I "), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), and granulocyte colony stimulating factor ("G-CSF")), or a growth factor (e.g., growth hormone ("GH")).
Techniques for conjugating such therapeutic moieties to antibodies are well known; see, e.g., Amon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al.
(eds.), 1985, pp. 243-56, Alan R. Liss, Inc.); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed), Robinson et al. (eds.), 1987, pp. 623-53, Marcel Dekker, Inc. ); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A
Review", in Monoclonal Antibodies `84: Biological And Clinical Applications, Pinchera et al. (eds.), 1985, pp. 475-506); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.),1985, pp. 303-16, Academic Press;
and Thorpe et al., Immunol. Recombinant expression vector., 62:119-58, 1982.
An antibody or fragment thereof, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
Alternatively, an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980.
Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.

5.4 METHODS OF PRODUCING FUSION PROTEINS
Fusion proteins can be produced by standard recombinant DNA techniques or by protein synthetic techniques, e.g., by use of a peptide synthesizer. For example, a nucleic acid molecule encoding a fusion protein can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Current Protocols in Molecular Biology, Ausubel et al., eds., John Wiley & Sons, 1992). Moreover, a nucleic acid encoding a bioactive molecule can be cloned into an expression vector containing the Fc domain or a fragment thereof such that the bioactive molecule is linked in-frame to the constant domain or fragment thereof.
Methods for fusing or conjugating polypeptides to the constant regions of antibodies are known in the art. See, e.g., U.S. Patent Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, 5,723,125, 5,783,181, 5,908,626, 5,844,095, and 5,112,946; EP
307,434; EP 367,166; EP 394,827; PCT publications WO 91/06570, WO 96/04388, WO
96/22024, WO 97/34631, and WO 99/04813; Ashkenazi et al., Proc. Natl. Acad.
Sci. USA, 88: 10535-10539, 1991; Traunecker et al., Nature, 331:84-86, 1988; Zheng et al., J.
Immunol., 154:5590-5600, 1995; and Vil et al., Proc. Natl. Acad. Sci. USA, 89:11337-11341, 1992.
The nucleotide sequence encoding a bioactive molecule may be obtained from any information available to those of skill in the art (e.g., from Genbank, the literature, or by routine cloning), and the nucleotide sequence encoding a constant domain or a fragment thereof with increased affinity for the FcRn may be determined by sequence analysis of mutants produced using techniques described herein, or may be obtained from Genbank or the literature. The nucleotide sequence coding for a fusion protein can be inserted into an appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted protein-coding sequence. A
variety of host-vector systems may be utilized in the present invention to express the protein-coding sequence. These include but are not limited to mammalian cell systems infected with virus (e.g., vaccinia virus, adenovirus, etc.); insect cell systems infected with virus (e.g., baculovirus); microorganisms such as yeast containing yeast vectors; or bacteria transformed with bacteriophage, DNA, plasmid DNA, or cosmid DNA. The expression elements of vectors vary in their strengths and specificities. Depending on the host-vector system utilized, any one of a number of suitable transcription and translation elements may be used.
The expression of a fusion protein may be controlled by any promoter or enhancer element known in the art. Promoters which may be used to control the expression of the gene encoding fusion protein include, but are not limited to, the SV40 early promoter region (Bemoist and Chambon, Nature, 290:304-310, 1981), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al., Cell, 22:787-797, 1980), the herpes thymidine kinase promoter (Wagner et al., Proc. Natl. Acad. Sci.
U.S.A., 78:1441-1445, 1981), the regulatory sequences of the metallothionein gene (Brinster et al., Nature, 296:39-42, 1982), the tetracycline (Tet) promoter (Gossen et al., Proc. Nat. Acad.
Sci. USA, 89:5547-5551, 1995); prokaryotic expression vectors such as the (3-lactamase promoter (Villa-Kamaroff, et al., Proc. Natl. Acad. Sci. U.S.A., 75:3727-3731, 1978), or the tac promoter (DeBoer, et al., Proc. Natl. Acad. Sci. U.S.A., 80:21-25, 1983;
see also "Useful proteins from recombinant bacteria" in Scientific American, 242:74-94, 1980);
plant expression vectors comprising the nopaline synthetase promoter region (Herrera-Estrella et al., Nature, 303:209-213, 1983) or the cauliflower mosaic virus 35S RNA
promoter (Gardner, et al., Nucl. Acids Res., 9:2871, 1981), and the promoter of the photosynthetic enzyme ribulose biphosphate carboxylase (Herrera-Estrella et al., Nature, 310:115-120, 1984); promoter elements from yeast or other fungi such as the Gal 4 promoter, the ADC
(alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, alkaline phosphatase promoter, and the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals:
elastase I gene control region which is active in pancreatic acinar cells (Swift et al., Cell 38:639-646, 1984; Ornitz et al., 50:399-409, Cold Spring Harbor Symp. Quant. Biol., 1986; MacDonald, Hepatology 7:425-515, 1987); insulin gene control region which is active in pancreatic beta cells (Harahan, Nature 315:115-122, 1985), immunoglobulin gene control region which is active in lymphoid cells (Grosschedl et al., Cell, 38:647-658, 1984; Adames et al., Nature 318:533-538, 1985; Alexander et al., Mol. Cell. Biol., 7:1436-1444, 1987), mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., Cell, 45:485-495, 1986), albumin gene control region which is active in liver (Pinkert et al., Genes and Devel., 1:268-276, 1987), a-fetoprotein gene control region which is active in liver (Krumlauf et al., Mol. Cell. Biol., 5:1639-1648, 1985;
Hammer et al., Science, 235:53-58, 1987; a 1-antitrypsin gene control region which is active in the liver (Kelsey et al., Genes and Devel., 1:161-171, 1987), beta-globin gene control region which is active in myeloid cells (Mogram et al., Nature, 315:338-340, 1985; Kollias et al., Cell, 46:89-94, 1986; myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al., Cell, 48:703-712, 1987); myosin light chain-2 gene control region which is active in skeletal muscle (Sani, Nature, 314:283-286, 1985);
neuronal-specific enolase (NSE) which is active in neuronal cells (Morelli et al., Gen. Virol., 80:571-83, 1999); brain-derived neurotrophic factor (BDNF) gene control region which is active in neuronal cells (Tabuchi et al., Biochem, Biophysic. Res.
Comprising., 253:818-823, 1998); glial fibrillary acidic protein (GFAP) promoter which is active in astrocytes (Gomes et al., Braz. J. Med. Biol. Res., 32(5):619-631, 1999; Morelli et at, Gen.
Virol., 80:571-83, 1999) and gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason et al., Science, 234:1372-1378, 1986).
In a specific embodiment, the expression of a fusion protein is regulated by a constitutive promoter. In another embodiment, the expression of a fusion protein is regulated by an inducible promoter. In accordance with these embodiments, the promoter may be a tissue-specific promoter.
In a specific embodiment, a vector is used that comprises a promoter operably linked to a fusion protein-encoding nucleic acid, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic resistance gene).
In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the fusion protein coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts (e.g., see Logan & Shenk, Proc. Natl. Acad. Sci. USA, 81:355-359,1984). Specific initiation signals may also be required for efficient translation of inserted fusion protein coding sequences. These signals include the ATG initiation codon and adjacent sequences.
Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al., Methods in Enzymol., 153:516-544, 1987).
Expression vectors containing inserts of a gene encoding a fusion protein can be identified by three general approaches: (a) nucleic acid hybridization, (b) presence or absence of "marker" gene functions, and (c) expression of inserted sequences.
In the first approach, the presence of a gene encoding a fusion protein in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous to an inserted gene encoding the fusion protein. In the second approach, the recombinant vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, etc.) caused by the insertion of a nucleotide sequence encoding a fusion protein in the vector. For example, if the nucleotide sequence encoding the fusion protein is inserted within the marker gene sequence of the vector, recombinants containing the gene encoding the fusion protein insert can be identified by the absence of the marker gene function. In the third approach, recombinant expression vectors can be identified by assaying the gene product (i.e., fusion protein) expressed by the recombinant. Such assays can be based, for example, on the physical or functional properties of the fusion protein in in vitro assay systems, e.g., binding with anti-bioactive molecule antibody.
In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus, expression of the genetically engineered fusion protein may be controlled.
Furthermore, different host cells have characteristic and specific mechanisms for the translational and post-translational processing and modification (e.g., glycosylation, phosphorylation of proteins). Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed. For example, expression in a bacterial system will produce an unglycosylated product and expression in yeast will produce a glycosylated product. Eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include but are not limited to CHO, VERY, BHK, HeLa, COS, MDCK, 293, 3T3, W138, and in particular, neuronal cell lines such as, for example, SK-N-AS, SK-N-FI, SK-N-DZ human neuroblastomas (Sugimoto et al., J Natl. Cancer Inst., 73: 51-57, 1984), SK-N-SH human neuroblastoma (Biochim.
Biophys. Acta, 704: 450-460, 1982), Daoy human cerebellar medulloblastoma (He et al., Cancer Res., 52: 1144-1148, 1992) DBTRG-05MG glioblastoma cells (Kruse et al., 1992, In Vitro Cell. Dev. Biol., 28A:609-614, 1992), IMR-32 human neuroblastoma (Cancer Res., 30:
2110-2118, 1970), 1321N1 human astrocytoma (Proc. Natl Acad. Sci. USA, 74:
4816, 1997), MOG-G-CCM human astrocytoma (Br. J. Cancer, 49: 269, 1984), U87MG human glioblastoma-astrocytoma (Acta Pathol. Microbiol. Scand., 74: 465-486, 1968), A172 human glioblastoma (Olopade et al., Cancer Res., 52: 2523-2529, 1992), C6 rat glioma cells (Benda et al., Science, 161: 370-371, 1968), Neuro-2a mouse neuroblastoma (Proc.
Natl. Acad. Sci.
USA, 65: 129-136, 1970), NB41A3 mouse neuroblastoma (Proc. Natl. Acad. Sci.
USA, 48:
1184-1190, 1962), SCP sheep choroid plexus (Bolin et al., J. Virol. Methods, 48: 211-221, 1994), G355-5, PG-4 Cat normal astrocyte (Haapala et al., J Virol., 53: 827-833, 1985), Mpf ferret brain (Trowbridge et al., In Vitro, 18: 952-960, 1982), and normal cell lines such as, for example, CTX TNA2 rat normal cortex brain (Radany et al., Proc. Natl.
Acad. Sci. USA, 89: 6467-6471, 1992) such as, for example, CRL7030 and Hs578Bst. Furthermore, different vector/host expression systems may effect processing reactions to different degrees.
For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the fusion protein may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched medium, and then are switched to a selective medium. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines that express the differentially expressed or pathway gene protein. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the differentially expressed or pathway gene protein.
A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler, et al., Cell, 11:223, 1997), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc.
Natl. Acad.
Sci. USA, 48:2026, 1962), and adenine phosphoribosyltransferase (Lowy, et al., 1980, Cell, 22:817, 1980) genes can be employed in tk-, hgprt- or aprt- cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler, et al., Natl. Acad. Sci. USA, 77:3567, 1980; O'Hare, et al., Proc. Natl. Acad. Sci. USA, 78:1527, 1981); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA, 78:2072, 1981); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin, et al., J. Mol.
Biol., 150:1, 1981);
and hygro, which confers resistance to hygromycin (Santerre, et al., Gene, 30:147, 1984) genes.
Once a fusion protein of the invention has been produced by recombinant expression, it may be purified by any method known in the art for purification of a protein, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.

5.5. PROPHYLACTIC AND THERAPEUTIC USES OF
ANTIBODIES

The present invention encompasses antibody-based therapies which involve administering antibodies to an animal, preferably a mammal, and most preferably a human, for preventing, treating, or ameliorating symptoms associated with a disease, disorder, or infection. Prophylactic and therapeutic compounds of the invention include, but are not limited to, antibodies and nucleic acids encoding antibodies. Antibodies may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
Antibodies of the present invention that function as antagonists of a disease, disorder, or infection can be administered to an animal, preferably a mammal and most preferably a human, to treat, prevent or ameliorate one or more symptoms associated with the disease, disorder, or infection. For example, antibodies which disrupt or prevent the interaction between a viral antigen and its host cell receptor may be administered to an animal, preferably a mammal and most preferably a human, to treat, prevent or ameliorate one or more symptoms associated with a viral infection.
In a specific embodiment, an antibody or fragment thereof prevents a viral or bacterial antigen from binding to its host cell receptor by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to antigen binding to its host cell receptor in the absence of said antibodies. In another embodiment, a combination of antibodies prevent a viral or bacterial antigen from binding to its host cell receptor by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to antigen binding to its host cell receptor in the absence of said antibodies. In a preferred embodiment, the antibody is used to treat or prevent RSV infection Antibodies which do not prevent a viral or bacterial antigen from binding its host cell receptor but inhibit or downregulate viral or bacterial replication can also be administered to an animal to treat, prevent or ameliorate one or more symptoms associated with a viral or bacterial infection. The ability of an antibody to inhibit or downregulate viral or bacterial replication may be determined by techniques described herein or otherwise known in the art. For example, the inhibition or downregulation of viral replication can be determined by detecting the viral titer in the animal.
In a specific embodiment, an antibody inhibits or downregulates viral or bacterial replication by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10%
relative to viral or bacterial replication in absence of said antibody. In another embodiment, a combination of antibodies inhibit or downregulate viral or bacterial replication by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to viral or bacterial replication in absence of said antibodies.
Antibodies can also be used to prevent, inhibit or reduce the growth or metastasis of cancerous cells. In a specific embodiment, an antibody inhibits or reduces the growth or metastasis of cancerous cells by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10%
relative to the growth or metastasis in absence of said antibody. In another embodiment, a combination of antibodies inhibits or reduces the growth or metastasis of cancer by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to the growth or metastasis in absence of said antibodies. Examples of cancers include, but are not limited to, leukemia (e.g., acute leukemia such as acute lymphocytic leukemia and acute myelocytic leukemia), neoplasms, tumors (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, and retinoblastoma), heavy chain disease, metastases, or any disease or disorder characterized by uncontrolled cell growth.

Antibodies can also be used to reduce the inflammation experienced by animals, particularly mammals, with inflammatory disorders. In a specific embodiment, an antibody reduces the inflammation in an animal by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to the inflammation in an animal in the not administered said antibody. In another embodiment, a combination of antibodies reduce the inflammation in an animal by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to the inflammation in an animal in not administered said antibodies. Examples of inflammatory disorders include, but are not limited to, rheumatoid arthritis, spondyloarthropathies, inflammatory bowel disease and asthma.
I5 In certain embodiments, the antibody used for treatment of inflammation (or cancer) is a modified anti-a,03 antibody, preferably a Vitaxin antibody (see, PCT publications WO 98/33919 and WO 00/78815, both by Huse et al.).

Antibodies can also be used to prevent the rejection of transplants.
Antibodies can also be used to prevent clot formation. Further, antibodies that function as agonists of the immune response can also be administered to an animal, preferably a mammal, and most preferably a human, to treat, prevent or ameliorate one or more symptoms associated with the disease, disorder, or infection.
One or more antibodies that immunospecifically bind to one or more antigens may be used locally or systemically in the body as a therapeutic. The antibodies of this invention may also be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), which, for example, serve to increase the number or activity of effector cells which interact with the antibodies. The antibodies of this invention may also be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), which, for example, serve to increase the immune response. The antibodies of this invention may also be advantageously utilized in combination with one or more drugs used to treat a disease, disorder, or infection such as, for example anti-cancer agents, anti-inflammatory agents or anti-viral agents. Examples of anti-cancer agents include, but are not limited to, isplatin, ifosfamide, paclitaxel, taxanes, topoisomerase I inhibitors (e.g., CPT-11, topotecan, 9-AC, and GG-21 1), gemcitabine, vinorelbine, oxaliplatin, 5-fluorouracil (5-FU), leucovorin, vinorelbine, temodal, and taxol. Examples of anti-viral agents include, but are not limited to, cytokines (e.g., IFN-a, IFN-0, IFN-y), inhibitors of reverse transcriptase (e.g., AZT, 3TC, D4T, ddC, ddl, d4T, 3TC, adefovir, efavirenz, delavirdine, nevirapine, abacavir, and other dideoxynucleosides or dideoxyfluoronucleosides), inhibitors of viral mRNA
capping, such as ribavirin, inhibitors of proteases such HIV protease inhibitors (e.g., amprenavir, indinavir, nelfinavir, ritonavir, and saquinavir,), amphotericin B, castanospermine as an inhibitor of glycoprotein processing, inhibitors of neuraminidase such as influenza virus neuraminidase inhibitors (e.g., zanamivir and oseltamivir), topoisomerase I inhibitors (e.g., camptothecins and analogs thereof), amantadine, and rimantadine. Examples of anti-inflammatory agents include, but are not limited to, nonsteroidal anti-inflammatory drugs such as inhibitors (e.g., meloxicam, celecoxib, rofecoxib, flosulide, and SC-58635, and MK-966), ibuprofen and indomethacin, and steroids (e.g., deflazacort, dexamethasone and methylprednisolone).
In a specific embodiment, antibodies administered to an animal are of a species origin or species reactivity that is the same species as that of the animal. Thus, in a preferred embodiment, human or humanized antibodies, or nucleic acids encoding human or human, are administered to a human patient for therapy or prophylaxis.
In preferred embodiments, immunoglobulins having extended in vivo half-lives are used in passive immunotherapy (for either therapy or prophylaxis).
Because of the extended half-life, passive immunotherapy or prophylaxis can be accomplished using lower doses and/or less frequent administration of the therapeutic resulting in fewer side effects, better patient compliance, less costly therapy/prophylaxis, etc. In a preferred embodiment, the therapeutic/prophylactic is an antibody that binds RSV, for example, SYNAGIS or other anti-RSV antibody. Such anti-RSV antibodies, and methods of administration are disclosed in U.S. patent application Serial Nos. 09/724,396 and 09/724,531, both entitled "Methods of Administering/Dosing Anti-RSV Antibodies For Prophylaxis and Treatment," both by Young et al., both filed November 28, 2000, and continuation-in-part applications of these applications, U.S. Patent Nos. 6,855,493 and 6,818,216, also entitled "Methods of Administering/Dosing Anti-RSV Antibodies for Prophylaxis and Treatment," by Young et al. Also included are the anti-RSV antibodies described in Section 5.1, supra.
In a specific embodiment, fusion proteins administered to an animal are of a species origin or species reactivity that is the same species is that of the animal. Thus, in a preferred embodiment, human fusion proteins or nucleic acids encoding human fusion proteins, are administered to a human subject for therapy or prophylaxis.

5.6. PROPHYLACTIC AND THERAPEUTIC
USES OF FUSION PROTEINS AND
CONJUGATED MOLECULES

The present invention encompasses fusion protein-based and conjugated molecule-based therapies which involve administering fusion proteins or conjugated molecules to an animal, preferably a mammal and most preferably a human, for preventing, treating, or ameliorating symptoms associated with a disease, disorder, or infection.
Prophylactic and therapeutic compounds of the invention include, but are not limited to, fusion proteins and nucleic acids encoding fusion proteins and conjugated molecules. Fusion proteins and conjugated molecules may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
Fusion proteins and conjugated molecules of the present invention that function as antagonists of a disease, disorder, or infection can be administered to an animal, preferably a mammal, and most preferably a human, to treat, prevent or ameliorate one or more symptoms associated with the disease, disorder, or infection. Further, fusion proteins and conjugated molecules of the present invention that function as agonists of the immune response may be administered to an animal, preferably a mammal, and most preferably a human, to treat, prevent or ameliorate one or more symptoms associated with the disease, disorder, or infection.
One or more fusion proteins and conjugated molecules may be used locally or systemically in the body as a therapeutic. The fusion proteins and conjugated molecules of this invention may also be advantageously utilized in combination with monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), which, for example, serve to increase the number or activity of effector cells which interact with the antibodies. The fusion proteins and conjugated molecules of this invention may also be advantageously utilized in combination with monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), which, for example, serve to increase the immune response.
The fusion proteins and conjugated molecules of this invention may also be advantageously utilized in combination with one or more drugs used to treat a disease, disorder, or infection such as, for example anti-cancer agents, anti-inflammatory agents or anti-viral agents.
Examples of anti-cancer agents include, but are not limited to, isplatin, ifosfamide, paclitaxel, taxanes, topoisomerase I inhibitors (e.g., CPT-11, topotecan, 9-AC, and GG-211), gemcitabine, vinorelbine, oxaliplatin, 5-fluorouracil (5-FU), leucovorin, vinorelbine, temodal, and taxol.
Examples of anti-viral agents include, but are not limited to, cytokines (e.g., IFN-a, IFN-0, IFN-y), inhibitors of reverse transcriptase (e.g., AZT, 3TC, D4T, ddC, ddl, d4T, 3TC, adefovir, efavirenz, delavirdine, nevirapine, abacavir, and other dideoxynucleosides or dideoxyfluoronucleosides), inhibitors of viral mRNA capping, such as ribavirin, inhibitors of proteases such HIV protease inhibitors (e.g., amprenavir, indinavir, nelfinavir, ritonavir, and saquinavir,), amphotericin B, castanospermine as an inhibitor of glycoprotein processing, inhibitors of neuraminidase such as influenza virus neuraminidase inhibitors (e.g., zanamivir and oseltamivir), topoisomerase I inhibitors (e.g., camptothecins and analogs thereof), amantadine, and rimantadine. Examples of anti-inflammatory agents include, but are not limted to, nonsteroidal anti-inflammatory drugs such as COX-2 inhibitors (e.g., meloxicam, celecoxib, rofecoxib, flosulide, and SC-58635, and MK-966), ibuprofen and indomethacin, and steroids (e.g., deflazacort, dexamethasone and methylprednisolone).

5.7. ADMINISTRATION OF ANTIBODIES OR FUSION
PROTEINS

The invention provides methods of treatment, prophylaxis, and amelioration of one or more symptoms associated with a disease, disorder or infection by administrating to a subject of an effective amount of an antibody of the invention, or pharmaceutical composition comprising an antibody of the invention. The invention also provides methods of treatment, prophylaxis, and amelioration of one or more symptoms associated with a disease, disorder or infection by administering to a subject an effective amount of a fusion protein or conjugated molecule of the invention, or a pharmaceutical composition comprising a fusion protein or conjugated molecules of the invention. In a preferred aspect, an antibody or fusion protein or conjugated molecule, is substantially purified (i.e., substantially free from substances that limit its effect or produce undesired side-effects). In a specific embodiment, the subject is an animal, preferably a mammal such as non-primate (e.g., cows, pigs, horses, cats, dogs, rats etc.) and a primate (e.g., monkey such as a cynomolgous monkey and a human). In a preferred embodiment, the subject is a human.
Various delivery systems are known and can be used to administer an antibody or fusion protein or conjugated molecule of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or fusion protein, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol.
Chem., 262:4429-4432, 1987), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of administering an antibody, a fusion protein or conjugated molecule, or pharmaceutical composition include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous), epidural, and mucosal (e.g., intranasal and oral routes). In a specific embodiment, antibodies, fusion proteins, conjugated molecules, or pharmaceutical compositions are administered intramuscularly, intravenously, or subcutaneously. The compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
Administration can be systemic or local. In addition, pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Patent Nos. 6,019,968; 5,985, 320; 5,985,309; 5,934,272; 5,874,064; 5,855,913;
5,290,540; and 4,880,078; and PCT Publication Nos. WO 92/19244; WO 97/32572; WO 97/44013; WO
98/31346; and WO 99/66903. In a preferred embodiment, an antibody, a fusion protein, conjugated molecules, or a pharmaceutical composition is administered using Alkermes AIRTM pulmonary drug delivery technology (Alkermes, Inc., Cambridge, MA).
The invention also provides that an antibody, a fusion protein, or conjugated molecule is packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of antibody, fusion protein, or conjugated molecule.
In one embodiment, the antibody, fusion protein, or conjugated molecule is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted, e.g., with water or saline to the appropriate concentration for administration to a subject. Preferably, the antibody, fusion protein, or conjugated molecule is supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more preferably at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, or at least 75 mg. The lyophilized antibody, fusion protein, or conjugated molecule should be stored at between 2 and 8 C in its original container and the antibody, fusion protein, or conjugated molecules should be administered within 12 hours, preferably within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted. In an alternative embodiment, an antibody, fusion protein, or conjugated molecule is supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the antibody, fusion protein, or conjugated molecule.

Preferably, the liquid form of the antibody, fusion protein, or conjugated molecule is supplied in a hermetically sealed container at least 1 mg/ml, more preferably at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/mI, at least 15 mg/kg, or at least 25 mg/ml.
In a specific embodiment, it may be desirable to administer the pharmaceutical compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
Preferably, when administering an antibody or a fusion protein, care must be taken to use materials to which the antibody or the fusion protein does not absorb.
In another embodiment, the composition can be delivered in a vesicle, in particular a liposome (see Langer, Science, 249:1527-1533, 1990; Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 3 17-327; see generally ibid.).
In yet another embodiment, the composition can be delivered in a controlled release or sustained release system. Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more antibodies, or one or more fusion proteins. See, e.g., U.S. Patent No. 4,526,938; PCT publication WO
91/05548;
PCT publication WO 96/20698; Ning et al., "Intratumoral Radioimmunotheraphy of a Human Colon Cancer Xenograft Using a Sustained-Release Gel," Radiotherapy &
Oncology, 39:179-189, 1996; Song et al., "Antibody Mediated Lung Targeting of Long-Circulating Emulsions," PDA Journal of Pharmaceutical Science & Technology, 50:372-397, 1995;
Cleek et al., "Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular Application," Pro. Intl. Symp. Control. Rel. Bioact. Mater., 24:853-854, 1997;
and Lam et al., "Microencapsulation of Recombinant Humanized Monoclonal Antibody for Local Delivery," Proc. Int'l. Symp. Control Rel. Bioact. Mater., 24:759-760, 1997.
In one embodiment, a pump may be used in a controlled release system (see Langer, supra;
Sefton, CRC Crit. Ref Biomed. Eng., 14:20, 1987; Buchwald et al., Surgery, 88:507, 1980; and Saudek et al., N. Engl. J. Med., 321:574, 1989). In another embodiment, polymeric materials can be used to achieve controlled release of antibodies or fusion proteins (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J Macromol. Sci. Rev. Macromol. Chem., 23:61, 1983;
see also Levy et al., Science, 228:190, 1985; During et al., Ann. Neurol., 25:351, 1989;

Howard et al., J Neurosurg., 7 1:105, 1989); U.S. Patent No. 5,679,377; U.S.
Patent No.
5,916,597; U.S. Patent No. 5,912,015; U.S. Patent No. 5,989,463; U.S. Patent No. 5,128,326;
PCT Publication No. WO 99/15154; and PCT Publication No. WO 99/20253). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target (e.g., the lungs), thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
Other controlled release systems are discussed in the review by Langer, Science, 249:1527-1533, 1990).
In a specific embodiment where the composition of the invention is a nucleic acid encoding an antibody or fusion protein, the nucleic acid can be administered in vivo to promote expression of its encoded antibody or fusion protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Patent No.
4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun;
Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see e.g., Joliot et al., Proc. Natl. Acad. Sci. USA, 88:1864-1868, 1991), etc. Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination.
The present invention also provides pharmaceutical compositions. Such compositions comprise a prophylactically or therapeutically effective amount of an antibody, fusion protein or conjugated molecule, and a pharmaceutically acceptable carrier. In a specific embodiment, the tern "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S.
Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant (e.g., Freund's complete and incomplete, mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful adjuvants for humans such as BCG
(Bacille Calmette-Guerin) and Corynebacterium parvum), excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin. Such compositions will contain a prophylactically or therapeutically effective amount of the antibody or fragment thereof, or fusion protein or conjugated molecule, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
In a preferred embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
Generally, the ingredients of compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
The compositions of the invention can be formulated as neutral or salt forms.
Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.

The amount of the composition of the invention which will be effective in the treatment, prevention or amelioration of one or more symptoms associated with a disease, disorder, or infection can be determined by standard clinical techniques. The precise dose to be employed in the formulation will depend on the route of administration, the age of the subject, and the seriousness of the disease, disorder, or infection, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model (e.g., the cotton rat or Cynomolgous monkey) test systems.
For fusion proteins, the therapeutically or prophylactically effective dosage administered to a subject ranges from about 0.001 to 50 mg/kg body weight, preferably about 0.01 to 25 mg/kg body weight, more preferably about 0.1 to 20 mg/kg body weight, and even more preferably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight. For antibodies, the therapeutically or prophylactically effective dosage administered to a subject is typically 0.1 mg/kg to 200 mg/kg of the subject's body weight.
Preferably, the dosage administered to a subject is between 0.1 mg/kg and 20 mg/kg of the subject's body weight and more preferably the dosage administered to a subject is between 1 mg/kg to 10 mg/kg of the subject's body weight. The dosage will, however, depend upon the extent to which the in vivo half-life of the molecule has been increased Generally, human antibodies and human fusion proteins have longer half-lives within the human body than antibodies of fusion proteins from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies or human fusion proteins and less frequent administration is often possible. Further, the dosage and frequency of administration of antibodies, fusion proteins, or conjugated molecules may be reduced also by enhancing uptake and tissue penetration (e.g., into the lung) of the antibodies or fusion proteins by modifications such as, for example, lipidation.
Treatment of a subject with a therapeutically or prophylactically effective amount of an antibody, fusion protein, or conjugated molecule can include a single treatment or, preferably, can include a series of treatments. In a preferred example, a subject is treated with an antibody, fusion protein, or conjugated molecule in the range of between about 0.1 to 30 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks. In other embodiments, the pharmaceutical composition of the invention is administered once a day, twice a day, or three times a day. In other embodiments, the pharmaceutical composition is administered once a week, twice a week, once every two weeks, once a month, once every six weeks, once every two months, twice a year or once per year. It will also be appreciated that the effective dosage of the antibody, fusion protein, or conjugated molecule used for treatment may increase or decrease over the course of a particular treatment.

5.7.1. GENE THERAPY
In a specific embodiment, nucleic acids comprising sequences encoding antibodies or fusion proteins, are administered to treat, prevent or ameliorate one or more symptoms associated with a disease, disorder, or infection, by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid. In this embodiment of the invention, the nucleic acids produce their encoded antibody or fusion protein that mediates a therapeutic or prophylactic effect.
Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described below.
For general reviews of the methods of gene therapy, see Goldspiel et al., Clinical Pharmacy, 12:488-505, 1993; Wu and Wu, Biotherapy, 3:87-95, 1991;
Tolstoshev, Ann. Rev. Pharmacol. Toxicol., 32:573-596, 1993; Mulligan, Science, 260:926-932, 1993;
and Morgan and Anderson, Ann. Rev. biochem. 62:191-217, 1993; TIBTECH
11(5):155-215, 1993. Methods commonly known in the art of recombinant DNA technology which can be used are described in Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993); and Kriegler, Gene Transfer and Expression, A
Laboratory Manual, Stockton Press, NY (1990).
In a preferred aspect, a composition of the invention comprises nucleic acids encoding an antibody, said nucleic acids being part of an expression vector that expresses the antibody in a suitable host. In particular, such nucleic acids have promoters, preferably heterologous promoters, operably linked to the antibody coding region, said promoter being inducible or constitutive, and, optionally, tissue-specific. In another particular embodiment, nucleic acid molecules are used in which the antibody coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the antibody encoding nucleic acids (Koller and Smithies, Proc. Natl. Acad. Sci. USA, 86:8932-8935, 1989; and Zijlstra et al., Nature, 342:435-438, 1989).
In another preferred aspect, a composition of the invention comprises nucleic acids encoding a fusion protein, said nucleic acids being a part of an expression vector that expression the fusion protein in a suitable host. In particular, such nucleic acids have promoters, preferably heterologous promoters, operably linked to the coding region of a fusion protein, said promoter being inducible or constitutive, and optionally, tissue-specific.
In another particular embodiment, nucleic acid molecules are used in which the coding sequence of the fusion protein and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the fusion protein encoding nucleic acids.
Delivery of the nucleic acids into a subject may be either direct, in which case the subject is directly exposed to the nucleic acid or nucleic acid-carrying vectors, or indirect, in which case, cells are first transformed with the nucleic acids in vitro, then transplanted into the subject. These two approaches are known, respectively, as in vivo or ex vivo gene therapy.
In a specific embodiment, the nucleic acid sequences are directly administered in vivo, where it is expressed to produce the encoded product. This can be accomplished by any of numerous methods known in the art, e.g., by constructing them as part of an appropriate nucleic acid expression vector and administering it so that they become intracellular, e.g., by infection using defective or attenuated retroviral or other viral vectors (see U.S. Patent No. 4,980,286), or by direct injection of naked DNA, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, encapsulation in liposomes, microparticles, or microcapsules, or by administering them in linkage to a peptide which is known to enter the nucleus, by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu and Wu, J Biol. Chem., 262:4429-4432, 1987) (which can be used to target cell types specifically expressing the receptors), etc. In another embodiment, nucleic acid-ligand complexes can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
In yet another embodiment, the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180; WO
92/22635;
WO 92/20316; WO 93/14188; WO 93/20221). Alternatively, the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, Proc. Natl. Acad. Sci. USA, 86:8932-8935, 1989; and Zijlstra et al., Nature, 342:435-438, 1989).
In a specific embodiment, viral vectors that contain nucleic acid sequences encoding an antibody or a fusion protein are used. For example, a retroviral vector can be used (see Miller et al., Meth. Enzymol., 217:581-599, 1993). These retroviral vectors contain the components necessary for the correct packaging of the viral genome and integration into the host cell DNA. The nucleic acid sequences encoding the antibody or a fusion protein to be used in gene therapy are cloned into one or more vectors, which facilitates delivery of the nucleotide sequence into a subject. More detail about retroviral vectors can be found in Boesen et al., Biotherapy, 6:291-302, 1994, which describes the use of a retroviral vector to deliver the mdr 1 gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy. Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., J. Clin. Invest., 93:644-651, 1994; Klein et al., Blood 83:1467-1473, 1994; Salmons and Gunzberg, Human Gene Therapy, 4:129-141, 1993; and Grossman and Wilson, Curr. Opin. in Genetics and Devel., 3:110-114, 1993.
Adenoviruses are other viral vectors that can be used in gene therapy.
Adenoviruses are especially attractive vehicles for delivering genes to respiratory epithelia.
Adenoviruses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson, Current Opinion in Genetics and Development, 3:499-503, 1993, present a review of adenovirus-based gene therapy. Bout et al., Human Gene Therapy, 5:3-10, 1994, demonstrated the use of adenovirus vectors to transfer genes to the respiratory epithelia of rhesus monkeys. Other instances of the use of adenoviruses in gene therapy can be found in Rosenfeld et al., Science, 252:431-434, 1991;
Rosenfeld et al., Cell, 68:143-155, 1992; Mastrangeli et al., J. Clin. Invest., 91:225-234, 1993; PCT
Publication WO 94/12649; and Wang et al., Gene Therapy, 2:775-783, 1995. In a preferred embodiment, adenovirus vectors are used.
Adeno-associated virus (AAV) has also been proposed for use in gene therapy (see, e.g.,Walsh et al., Proc. Soc. Exp. Biol. Med., 204:289-300, 1993, and U.S. Patent No.
5,436,146).
Another approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection. Usually, the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a subject.
In this embodiment, the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell. Such introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcellmediated gene transfer, spheroplast fusion, etc. Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Loeffler and Behr, Meth. Enzymol., 217:599-618, 1993;
Cohen et al., Meth. Enzymol., 217:618-644, 1993; and Clin. Pharma. They., 29:69-92, 1985) and may be used in accordance with the present invention, provided that the necessary developmental and physiological functions of the recipient cells are not disrupted. The technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
The resulting recombinant cells can be delivered to a subject by various methods known in the art. Recombinant blood cells (e.g., hematopoietic stem or progenitor cells) are preferably administered intravenously. The amount of cells envisioned for use depends on the desired effect, patient state, etc., and can be determined by one skilled in the art.
Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes;
blood cells such as T
lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood, fetal liver, etc.
In a preferred embodiment, the cell used for gene therapy is autologous to the subject.
In an embodiment in which recombinant cells are used in gene therapy, nucleic acid sequences encoding an antibody or a fusion protein are introduced into the cells such that they are expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect. In a specific embodiment, stem or progenitor cells are used. Any stem and/or progenitor cells which can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention (see e.g., PCT Publication WO 94/08598; Stemple and Anderson, Cell, 7 1:973-985, 1992;
Rheinwald, Meth. Cell Bio., 21A:229, 1980; and Pittelkow and Scott, Mayo Clinic Proc., 61:771, 1986).
In a specific embodiment, the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding region, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.

5.8. CHARACTERIZATION AND DEMONSTRATION OF
THERAPEUTIC OR PROPHYLACTIC UTILITY
Antibodies, fusion proteins, and conjugated molecules of the present invention may be characterized in a variety of ways. In particular, antibodies of the invention may be assayed for the ability to immunospecifically bind to an antigen. Such an assay may be performed in solution (e.g., Houghten, BiolTechniques, 13:412-421, 1992), on beads (Lam, Nature, 354:82-84, 1991, on chips (Fodor, Nature, 364:555-556, 1993), on bacteria (U.S. Patent No. 5,223,409), on spores (U.S. Patent Nos. 5,571,698; 5,403,484;
and 5,223,409), on plasmids (Cull et al., Proc. Natl. Acad. Sci. USA, 89:1865-1869, 1992) or on phage (Scott and Smith, Science, 249:386-390, 1990; Devlin, Science, 249:404-406, 1990;
Cwirla et al., Proc. Natl. Acad Sci. USA, 87:6378-6382,1990; and Felici, J.
Mol. Biol., 222:301-310, 1991). Antibodies that have been identified to immunospecitically bind to an antigen or a fragment thereof can then be assayed for their specificity affinity for the antigen.
The antibodies of the invention or fragments thereof may be assayed for immunospecific binding to an antigen and cross-reactivity with other antigens by any method known in the art. Immunoassays which can be used to analyze immunospecific binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA
(enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol.
1, John Wiley &
Sons, Inc., New York). Exemplary immunoassays are described briefly below (but are not intended by way of limitation).
Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the antibody of interest to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 C, adding protein A and/or protein G sepharose beads to the cell lysate, incubating for about an hour or more at 40 C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. The ability of the antibody of interest to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody to an antigen and decrease the background (e.g., pre-y clearing the cell lysate with sepharose beads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1.
Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20%
SDS-PAGE
depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), blocking the membrane with primary antibody (the antibody of interest) diluted in blocking buffer, washing the membrane in washing buffer, blocking the membrane with a secondary antibody (which recognizes the primary antibody, e.g., an anti-human antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32P or 125I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
ELISAs comprise preparing antigen, coating the well of a 96 well microtiter plate with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen. In ELISAs the antibody of interest does not have to be conjugated to a detectable compound;
instead, a second antibody (which recognizes the antibody of interest) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, the antibody may be coated to the well. In this case, a second antibody conjugated to a detectable compound may be added following the addition of the antigen of interest to the coated well. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1.

The binding affinity of an antibody to an antigen and the off-rate of an antibody-antigen interaction can be determined by competitive binding assays.
One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 125I) with the antibody of interest in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen. The affinity of the antibody of the present invention or a fragment thereof for the antigen and the binding off-rates can be determined from the saturation data by scatchard analysis.
Competition with a second antibody can also be determined using radioimmunoassays. In this case, the antigen is incubated with an antibody of the present invention or a fragment thereof conjugated to a labeled compound (e.g., 3H or 125I) in the presence of increasing amounts of an unlabeled second antibody.
In a preferred embodiment, BlAcore kinetic analysis is used to determine the binding on and off rates of antibodies to an antigen. BlAcore kinetic analysis comprises analyzing the binding and dissociation of an antigen from chips with immobilized antibodies on their surface (see the Example section infra).
The antibodies of the invention as well as fusion proteins and conjugated molecules can also be assayed for their ability to inhibit the binding of an antigen to its host cell receptor using techniques known to those of skill in the art. For example, cells expressing the receptor for a viral antigen can be contacted with virus in the presence or absence of an antibody and the ability of the antibody to inhibit viral antigen's binding can measured by, for example, flow cytometry or a scintillation counter. The antigen or the antibody can be labeled with a detectable compound such as a radioactive label (e.g., 32P, 355, and 125I) or a fluorescent label (e.g., fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine) to enable detection of an interaction between the antigen and its host cell receptor.
Alternatively, the ability of antibodies to inhibit an antigen from binding to its receptor can be determined in cell-free assays. For example, virus or a viral antigen (e.g., RSV F
glycoprotein) can be contacted in a cell-free assay with an antibody and the ability of the antibody to inhibit the virus or the viral antigen from binding to its host cell receptor can be determined. Preferably, the antibody is immobilized on a solid support and the antigen is labeled with a detectable compound. Alternatively, the antigen is immobilized on a solid support and the antibody is labeled with a detectable compound. The antigen may be partially or completely purified (e.g., partially or completely free of other polypeptides) or part of a cell lysate. Further, the antigen may be a fusion protein comprising the viral antigen and a domain such as glutathionine-S-transferase. Alternatively, an antigen can be biotinylated using techniques well known to those of skill in the art (e.g., biotinylation kit, Pierce Chemicals; Rockford, IL).
The antibodies, fusion proteins, and conjugated molecules of the invention can also be assayed for their ability to inhibit or downregulate viral or bacterial replication using techniques known to those of skill in the art. For example, viral replication can be assayed by a plaque assay such as described, e.g., by Johnson et al., Journal of Infectious Diseases, 176:1215-1224, 1997. The antibodies, fusion proteins, and conjugated molecules of the invention of the invention can also be assayed for their ability to inhibit or downregulate the expression of viral or bacterial polypeptides. Techniques known to those of skill in the art, including, but not limited to, Western blot analysis, Northern blot analysis, and RT-PCR, can be used to measure the expression of viral or bacterial polypeptides.
Further, the antibodies, fusion proteins, and conjugated molecules of the invention of the invention can be assayed for their ability to prevent the formation of syncytia.
The antibodies, fusion proteins, conjugated molecules, and compositions of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans. For example, in vitro assays which can be used to determine whether administration of a specific antibody, a specific fusion protein, a specific conjugated molecule, or a composition of the present invention is indicated, include in vitro cell culture assays in which a subject tissue sample is grown in culture, and exposed to or otherwise administered an antibody, a fusion protein, conjugated molecule, or composition of the present invention, and the effect of such an antibody, a fusion protein, conjugated molecule, or a composition of the present invention upon the tissue sample is observed. In various specific embodiments, in vitro assays can be carried out with representative cells of cell types involved in a disease or disorder, to determine if an antibody, a fusion protein, conjugated molecule, or composition of the present invention has a desired effect upon such cell types. Preferably, the antibodies, the fusion proteins, the conjugated molecules, or compositions of the invention are also tested in in vitro assays and animal model systems prior to administration to humans.
Antibodies, fusion proteins, conjugated molecules, or compositions of the present invention for use in therapy can be tested for their toxicity in suitable animal model systems, including but not limited to rats, mice, cows, monkeys, and rabbits.
For in vivo testing for the toxicity of an antibody, a fusion protein, a conjugated molecule, or a composition, any animal model system known in the art may be used.
Efficacy in treating or preventing viral infection may be demonstrated by detecting the ability of an antibody, a fusion protein, a conjugated molecule, or a composition of the invention to inhibit the replication of the virus, to inhibit transmission or prevent the virus from establishing itself in its host, or to prevent, ameliorate or alleviate one or more symptoms associated with viral infection. The treatment is considered therapeutic if there is, for example, a reduction is viral load, amelioration of one or more symptoms or a decrease in mortality and/or morbidity following administration of an antibody, a fusion protein, a conjugated molecule, or a composition of the invention. Antibodies, fusion proteins, conjugated molecules, or compositions of the invention can also be tested for their ability to inhibit viral replication or reduce viral load in in vitro and in vivo assays.
Efficacy in treating or preventing bacterial infection may be demonstrated by detecting the ability of an antibody, a fusion protein or a composition of the invention to inhibit the bacterial replication, or to prevent, ameliorate or alleviate one or more symptoms associated with bacterial infection. The treatment is considered therapeutic if there is, for example, a reduction is bacterial numbers, amelioration of one or more symptoms or a decrease in mortality and/or morbidity following administration of an antibody, a fusion protein or a composition of the invention.
Efficacy in treating cancer may be demonstrated by detecting the ability of an antibody, a fusion protein, a conjugated molecule, or a composition of the invention to inhibit or reduce the growth or metastasis of cancerous cells or to ameliorate or alleviate one or more symptoms associated with cancer. The treatment is considered therapeutic if there is, for example, a reduction in the growth or metastasis of cancerous cells, amelioration of one or more symptoms associated with cancer, or a decrease in mortality and/or morbidity following administration of an antibody, a fusion protein, a conjugated molecule, or a composition of the invention. Antibodies, fusion proteins or compositions of the invention can be tested for their ability to reduce tumor formation in in vitro, ex vivo, and in vivo assays.
Efficacy in treating inflammatory disorders may be demonstrated by detecting the ability of an antibody, a fusion protein, a conjugated molecule, or a composition of the invention to reduce or inhibit the inflammation in an animal or to ameliorate or alleviate one or more symptoms associated with an inflammatory disorder. The treatment is considered therapeutic if there is, for example, a reduction is in inflammation or amelioration of one or more symptoms following administration of an antibody, a fusion proteins, a conjugated molecule, or a composition of the invention.
Antibodies, fusion proteins, conjugated molecules, or compositions of the invention can be tested in vitro and in vivo for the ability to induce the expression of cytokines (e.g., IFN-a, IFN-(3, IFN-y, IL-2, IL-3, IL-4, IL-5, IL-6, IL10, IL-
12, and IL-15) and activation markers (e.g., CD28, ICOS, and SLAM). Techniques known to those of skill in the art can be used to measure the level of expression of cytokines and activation markers.
For example, the level of expression of cytokines can be measured by analyzing the level of RNA of cytokines by, for example, RT-PCR and Northern blot analysis, and by analyzing the level of cytokines by, for example, immunoprecipitation followed by Western blot analysis or ELISA.
Antibodies, fusion proteins, conjugated molecules, or compositions of the invention can be tested in vitro and in vivo for their ability to modulate the biological activity of immune cells, preferably human immune cells (e.g., T-cells, B-cells, and Natural Killer cells). The ability of an antibody, a fusion protein, a conjugated molecule, or a composition of the invention to modulate the biological activity of immune cells can be assessed by detecting the expression of antigens, detecting the proliferation of immune cells, detecting the activation of signaling molecules, detecting the effector function of immune cells, or detecting the differentiation of immune cells. Techniques known to those of skill in the art can be used for measuring these activities. For example, cellular proliferation can be assayed by 3H-thymidine incorporation assays and trypan blue cell counts. Antigen expression can be assayed, for example, by immunoassays including, but are not limited to, competitive and non-competitive assay systems using techniques such as Western blots, immunohistochemistry, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A
immunoassays and FACS analysis. The activation of signaling molecules can be assayed, for example, by kinase assays and electrophoretic shift assays (EMSAs).
Antibodies, fusion proteins, conjugated molecules, or compositions of the invention can also be tested for their ability to increase the survival period of animals, preferably mammals and most preferably humans, suffering from a disease, disorder, or infection by at least 25%, preferably at least 50%, at least 60%, at least 75%, at least 85%, at least 95%, or at least 99%. Further, antibodies, fusion proteins, conjugated molecules, or compositions of the invention can be tested for their ability reduce the hospitalization period of animals, preferably mammals and most preferably humans, suffering from a disease, disorder, or infection by at least 60%, preferably at least 75%, at least 85%, at least 95%, or at least 99%. Techniques known to those of skill in the art can be used to analyze the function of the antibodies or compositions of the invention in vivo.

5.9. DIAGNOSTIC USES OF ANTIBODIES AND FUSION
PROTEINS

Labeled antibodies, fusion proteins, and conjugated molecules of the invention can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders or infections. The invention provides for the detection or diagnosis of a disease, disorder or infection, comprising: (a) assaying the expression of an antigen in cells or a tissue sample of a subject using one or more antibodies that immunospecifically bind to the antigen; and (b) comparing the level of the antigen with a control level, e.g., levels in normal tissue samples, whereby an increase in the assayed level of antigen compared to the control level of the antigen is indicative of the disease, disorder or infection. The invention also provides for the detection or diagnosis of a disease, disorder or infection, comprising (a) assaying the expression of an antigen in cells or a tissue sample of a subject using one or fusion proteins or conjugated molecules of the invention that bind to the antigen; and (b) comparing the level of the antigen with a control level, e.g., levels in normal tissue samples, whereby an increase of antigen compared to the control level of the antigen is indicative of the disease, disorder or infection. Accordingly, the fusion protein or conjugated molecule comprises a bioactive molecule such as a ligand, cytokine or growth factor and the hinge-Fc region or fragments thereof, wherein the fusion protein or conjugated molecule is capable of binding to an antigen being detected.
Antibodies of the invention can be used to assay antigen levels in a biological sample using classical immunohistological methods as described herein or as known to those of skill in the art (e.g., see Jalkanen et al., J. Cell. Biol., 101:976-985, 1985; Jalkanen et al., J Cell. Biol., 105:3087-3096, 1987). Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, alkaline phosphatase, glucose oxidase;
radioisotopes, such as iodine (251, 1311)' carbon (14C), sulfur (35S), tritium (3H), indium (21In), and technetium (99mTc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine.
Fusion proteins can be used to assay antigen levels in a biological sample using, for example, SDS-PAGE and immunoassays known to those of skill in the art.
One aspect of the invention is the detection and diagnosis of a disease, disorder, or infection in a human. In one embodiment, diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled antibody that immunospecifically binds to an antigen; b) waiting for a time interval following the administration for permitting the labeled antibody to preferentially concentrate at sites in the subject where the antigen is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled antibody in the subject, such that detection of labeled antibody above the background level indicates that the subject has the disease, disorder, or infection. In accordance with this embodiment, the antibody is labeled with an imaging moiety which is detectable using an imaging system known to one of skill in the art.
Background level can be determined by various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system.
In another embodiment, diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled fusion protein or conjugated molecule that binds to an antigen or some other molecule; b) waiting for a time interval following the administration for permitting the labeled fusion protein or conjugated molecule to preferentially concentrate at sites in the subject where the antigen or other molecule is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled fusion protein or conjugated molecule in the subject, such that detection of labeled fusion protein above the background level indicates that the subject has the disease, disorder, or infection. In accordance with this embodiment, the fusion protein or conjugated molecule comprises a bioactive molecule such as a ligand, cytokine or growth factor and a hinge-Fc region or a fragment thereof, wherein said fusion protein or conjugated molecule is labeled with an imaging moiety and is capable of binding to the antigen being detected.
It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. The labeled antibody will then preferentially accumulate at the location of cells which contain the specific protein. In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments,"
Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B. A.
Rhodes, eds., Masson Publishing Inc. (1982).
Depending on several variables, including the type of label used and the mode of administration, the time interval following the administration for permitting the labeled molecule to preferentially concentrate at sites in the subject and for unbound labeled molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours or 6 to 12 hours.

In another embodiment the time interval following administration is 5 to 20 days or 5 to 10 days.
In one embodiment, monitoring of a disease, disorder or infection is carried out by repeating the method for diagnosing the disease, disorder or infection, for example, one month after initial diagnosis, six months after initial diagnosis, one year after initial diagnosis, etc.
Presence of the labeled molecule can be detected in the subject using methods known in the art for in vivo scanning. These methods depend upon the type of label used.
Skilled artisans will be able to determine the appropriate method for detecting a particular label. Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
In a specific embodiment, the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurston et al., U.S.
Patent No. 5,441,050). In another embodiment, the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument.
In another embodiment, the molecule is labeled with a positron emitting metal and is detected in the patient using positron emission-tomography. In yet another embodiment, the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI).

5.10. KITS
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
The present invention provides kits that can be used in the above methods. In one embodiment, a kit comprises an antibody, fusion protein, or conjugated molecule, of the invention, preferably in a purified form, in one or more containers. In a specific embodiment, the kits of the present invention contain a substantially isolated antigen as a control. Preferably, the kits of the present invention further comprise a control antibody, fusion protein, or conjugated molecule which does not react with the antigen included in the kit. In another specific embodiment, the kits of the present invention contain a means for detecting the binding of an antibody, fusion protein, or conjugated molecule, to an antigen (e.g., the antibody, fusion protein, or conjugated molecule, may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate). In specific embodiments, the kit may include a recombinantly produced or chemically synthesized antigen. The antigen provided in the kit may also be attached to a solid support. In a more specific embodiment the detecting means of the above-described kit includes a solid support to which antigen is attached. Such a kit may also include a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the antigen can be detected by binding of the said reporter-labeled antibody.

5.11 IN VITRO AND IN VIVO ASSAYS FOR EXTENDED
HALF-LIFE OF MODIFIED IGG HINGE-FC
FRAGMENTS
The binding ability of modified IgGs and molecules comprising an IgG
constant domain of FcRn fragment thereof to FeRn can be characterized by various in vitro assays. PCT publication WO 97/34631 by Ward discloses various methods in detail.

For example, in order to compare the ability of the modified IgG or fragments thereof to bind to FcRn with that of the wild type IgG, the modified IgG or fragments thereof and the wild type IgG can be radio-labeled and reacted with FcRn-expressing cells in vitro.
The radioactivity of the cell-bound fractions can be then counted and compared. The cells expressing FcRn to be used for this assay are preferably endothelial cell lines including mouse pulmonary capillary endothelial cells (B 10, D2.PCE) derived from lungs of B10.DBA/2 mice and SV40 transformed endothelial cells (SVEC) (Kim et al., .1.
Immunol., 40:457-465, 1994) derived from C3H/HeJ mice. However, other types of cells, such as intestinal brush borders isolated from 10- to 14-day old suckling mice, which express sufficient number of FcRn can be also used. Alternatively, mammalian cells which express recombinant FcRn of a species of choice can be also utilized. After counting the radioactivity of the bound fraction of modified IgG or that of wild type, the bound molecules can be then extracted with the detergent, and the percent release per unit number of cells can be calculated and compared.
Affinity of modified IgGs for FcRn can be measured by surface plasmon resonance (SPR) measurement using, for example, a BlAcore 2000 (BlAcore Inc.) as described previously (Popov et al., Mol. Immunol., 33:493-502, 1996; Karlsson et al., J
Immunol. Methods, 145:229-240, 1991). In this method, FeRn molecules are coupled to a BlAcore sensor chip (e.g., CM5 chip by Pharmacia) and the binding of modified IgG
to the immobilized FcRn is measured at a certain flow rate to obtain sensorgrams using BIA evaluation 2.1 software, based on which on- and off-rates of the modified IgG, constant domains, or fragments thereof, to FcRn can be calculated.

Relative affinities of modified IgGs or fragments thereof, and the wild type IgG for FcRn can be also measured by a simple competition binding assay.
Unlabeled modified IgG or wild type 1gG is added in different amounts to the wells of a 96-well plate in which FcRn is immobilize. A constant amount of radio-labeled wild type IgG is then added to each well. Percent radioactivity of the bound fraction is plotted against the amount of unlabeled modified IgG or wild type IgG and the relative affinity of the modified hinge-Fe can be calculated from the slope of the curve.
Furthermore, affinities of modified IgGs or fragments thereof, and the wild type IgG for FcRn can be also measured by a saturation study and the Scatchard analysis.
Transfer of modified IgG or fragments thereof across the cell by FeRn can be measured by in vitro transfer assay using radiolabeled IgG or fragments thereof and FcRn-expressing cells and comparing the radioactivity of the one side of the cell monolayer with that of the other side. Alternatively, such transfer can be measured in vivo by feeding 10- to 14-day old suckling mice with radiolabeled, modified IgG and periodically counting the radioactivity in blood samples which indicates the transfer of the IgG through the intestine to the circulation (or any other target tissue, e.g., the lungs). To test the dose-dependent inhibition of the IgG transfer through the gut, a mixture of radiolabeled and unlabeled IgG at certain ratio is given to the mice and the radioactivity of the plasma can be periodically measured (Kim et al., Eur. J. Immunol., 24:2429-2434, 1994).
The half-life of modified IgG or fragments thereof can be measure by pharmacokinetic studies according to the method described by Kim et al. (Eur.
J ofImmuno.
24:542, 1994). According to this method, radiolabeled modified IgG or fragments thereof is injected intravenously into mice and its plasma concentration is periodically measured as a function of time, for example, at 3 minutes to 72 hours after the injection.
The clearance curve thus obtained should be biphasic, that is, a-phase and p-phase. For the determination of the in vivo half-life of the modified IgGs or fragments thereof, the clearance rate in 0-phase is calculated and compared with that of the wild type IgG.

6. EXAMPLES
The following examples illustrate the production, isolation, and characterization of modified hinge-Fc fragments that have longer in vivo half-lives.
6.1 LIBRARY CONSTRUCTION
6.1.1 REAGENTS
All chemicals were of analytical grade. Restriction enzymes and DNA-modifying enzymes were purchased from New England Biolabs, Inc. (Beverly, MA).
Oligonucleotides were synthesized by MWG Biotech, Inc. (High Point, NC).
pCANTAB5E
phagemid vector, anti-E-tag-horseradish peroxydase conjugate, TG1 E. Coli strain, IgG
Sepharose 6 Fast Flow and HiTrap protein A columns were purchased from APBiotech, Inc.
(Piscataway, NJ). VCSM13 helper phage and the Quick change mutagenesis kit were obtained from Stratagene (La Jolla, CA). CJ236 E. coli strain was purchased from Bio-Rad (Richmond, CA). BCA Protein Assay Reagent Kit was obtained from Pierce (Rockford, IL).
Lipofectamine 2000 was purchased from Invitrogen, Inc. (Carlsbad, CA).

6.1.2 EXPRESSION AND PURIFICATION OF
MURINE AND HUMAN FCRN

The amino acid sequences of human and mouse FcRn are SEQ ID NOs. 84 and 85, respectively (see also Firan et at, Intern. Immunol., 13:993-1002, 2001 and Popov et at, Mol. Immunol., 33:521-530, 1996). Human FcRn was also obtained following isolation from human placenta cDNA (Clontech, Palo Alto, CA) of the genes for human R2-microglobulin (Kabat et at, 1991, Sequences of Proteins of Immunological Interest, U.S. Public Health Service, National Institutes of Health, Washington, DC) and codons -23 to 267 of the human a chain (Story et al., J. Exp. Med., 180:2377-2381, 1994) using standard PCR protocols. Light and heavy chains along with their native signal sequence (Kabat et al., 1991, supra; Story et al., supra) were cloned in pFastBac DUAL
and pFastBac 1 bacmids, respectively, and viral stocks produced in Spodopterafrugiperda cells (Sf9) according to the manufacturer's instructions (Invitrogen, Carlsbad, CA).
High-Five cells were infected at a multiplicity of infection of 3 with the baculoviruses encoding a and (32 chains using commercially available protocols (Invitrogen).
Recombinant human FcRn was purified as follows: supernatant of infected insect cells was dialyzed into 50 mM MES (2-N-[Morpholino]ethansulfonic acid) pH 6.0 and applied to a 10 ml human IgG Sepharose 6 Fast Flow column (APBiotech, Piscataway, NJ).
Resin was washed with 200 ml 50 mM MES pH 6.0 and FcRn eluted with 0.1 M Tris-Cl pH
8Ø Purified FcRn was dialyzed against 50 mM MES pH 6.0, flash frozen and stored at -70 C. The purity of proteins was checked by SDS-PAGE and HPLC.

6.1.3 PREPARATION OF TAA-CONTAINING ssDNA
URACIL TEMPLATE

Construction of the libraries was based on a site directed mutagenesis strategy derived from the Kunkel method (Kunkel et al., Methods Enzymol. 154:367-382, 1987). A
human hinge-Fc gene spanning amino acid residues 226-478 (Kabat numbering, Kabat et al., 1991, supra) derived from MEDI-493 human IgG1 (Johnson et al., J Infect.
Disease, 176:1215-1224, 1997), was cloned into the pCANTAB5E phagemid vector as an Sfil/Notl fragment. Four libraries were generated by introducing random mutations at positions 251, 252, 254, 255, 256 (library 1), 308, 309, 311, 312, 314 (library 2), 385, 386, 387, 389 (library 3) and 428, 433, 434, 436 (library 4). Briefly, four distinct hinge-Fc templates were generated using PCR by overlap extension (Ho et al., Gene, 15:51-59, 1989), each containing one TAA stop codon at position 252 (library 1), 310 (library 2), 384 (library 3) or 429 (library 4), so that only mutagenized phagemids will give rise to Fc-displaying phage.
Each TAA-containing single-stranded DNA (TAAssDNA) was then prepared as follows: a single CJ236 E. coli colony harboring one of the four relevant TAA-containing phagemids was grown in 10 ml 2 x YT medium supplemented with 10 g/ml chloramphenicol and 100 g/ml ampicillin. At OD600 =1, VCSM 13 helper phage was added to a final concentration of 1010 pfu/ml. After 2 hours, the culture was transferred to 500 ml of 2 x YT medium supplemented with 0.25 g/ml uridine, 10 g/ml chloramphenicol, 30 g/ nl kanamycin, 100 g/ml ampicillin and grown overnight at 37 C. Phage were precipitated with PEG6000 using standard protocols (Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, New York, Vols. 1-3) and purified using the Qiaprep Spin M13 Kit (Qiagen, Valencia, CA) according to the manufacturer's instructions. 10 to 30 g of each uracil-containing TAAssDNA template was then combined with 0.6 g of the following phosphorylated oligonucleotides (randomized regions underlined) in 50 mM Tris-HCl, 10 mM MgC12, pH 7.5 in a final volume of 250 l:
Library 1:
5'-CATGTGACCTCAGGSNNSNNSNNGATSNNSNNGGTGTCCTTGGGTTTT

GGGGGG-3' (SEQ ID NO:120) Library 2:
5'-GCACTTGTACTCCTTGCCATTSNNCCASNNSNNGTGSNNSNNGGTGA
GGACGC-3' (SEQ ID NO:121) Library 3:
5'-GGTCTTGTAGTTSNNCTCSNNSNNSNNATTGCTCTCCC-3' (SEQ ID NO:122) Library 4:
5'-GGCTCTTCTGCGTSNNGTGSNNSNNCAGAGCCTCATGSNNCACGGAGC
ATGAG-3' (SEQ ID NO:123) whereN=A, C, Tor GandS = GorC.

6.1.4 SYNTHESIS OF HETERODUPLEX DNA
Appropriate, degenerate oligonucleotides were phosphorylated in the presence of T4 polynucleotide kinase using the standard protocol. Ten to 30 g of ssDNA
U template and 0.6 g of phosphorylated oligonucleotide were combined in 50 mM Tris-HCl containing 10 mM MgCl2, pH 7.5, to a final volume of 250 l and incubated at 90 C for 2 minutes, 50 C
for 3 minutes, and 20 C for 5 minutes. Synthesis of the heteroduplex DNA was carried out by adding 30 units of both T4 DNA ligase and T7 DNA polyinerase in the presence of 0.4 mM ATP, 1 mM dNTPs and 6 mM DTT and the mixture was incubated for 4 hours at 20 C.
The heteroduplex DNA thus produced was then purified and desalted using Qiagen Qiaquick DNA purification Kit (Qiagen, CA).

6.1.5 ELECTROPORATION
300 l electrocompetent TG1 E. coli cells were electroporated with 1 to 5 .ig of the heteroduplex DNA in a 2.5 kV field using 200 SZ and 25 F capacitance until a library size of 1 x 108 (library 1 and 2) or 1 x 10' (library 3 and 4) was reached.
The cells were resuspended in 2 ml SOC medium and the procedure was repeated 6 to 10 times.
The diversity was assessed by titration of recombinant E. coli. The pulsed cells were incubated in 50 ml SOC medium for 30 minutes at 37 C under agitation, centrifuged, and resuspended in 500 ml 2xYT containing 100 g/ml ampicillin and 1010 pfu/ml of VCSM13 helper phage.
The culture was incubated overnight at 37 C and the cells were pelleted by centrifugation.
The phage in the supernatant which express mutated hinge-Fc portion on its GIII-coat protein were precipitated with PEG6000 as previously described (Sambrook et al., 1989, supra) and resuspended in 5 ml of 20 mM MES, pH 6Ø

6.2 PANNING OF THE LIBRARY
Phage were panned using an ELISA-based approach. A 96-well ELISA plate was coated with 100 l/well of 0.01 mg/ml murine FcRn in sodium carbonate buffer, pH
9.0, at 4 C overnight and then blocked with 4% skimmed milk at 37 C for 2 hours. In each well of the coated plate, 100-150 l of the phage suspension (about 1013 phage in total) in 20 mM MES, pH 6.0, containing 5% milk and 0.05% Tween 20, were placed and incubated at 37 C for two to three hours with agitation.
After the incubation, the wells were washed with 20 mM MES, pH 6.0, containing 0.2% Tween 20 and 0.3 M NaCl about thirty times at room temperature. The bound phage were eluted with 100 l/well of PBS, pH 7.4, at 37 C for 30 minutes.
The eluted phage were then added to the culture of exponentially growing E.
coli cells and propagation was carried out overnight at 37 C in 250 ml 2xYT
supplemented with 100 4g/ml ampicillin and 1010 pfu/ml of VCSM13 helper phage. Propagated phage were collected by centrifugation followed by precipitation with PEG and the panning process was repeated up to a total of six times.
For the phage library containing mutations in residues 308-314 (H310 and W313 fixed), the phage expressing hinge-Fc region with higher affinities for FcRn were enriched by each panning process as shown in Table IV. The panning results of the library for the mutations in the residues 251-256 (1253 fixed) and that of the library for the mutations in the residues 428-436 (H429, E430, A431, L432, and H435 fixed), are shown in Tables V and VI, respectively. Furthermore, the panning results of the library for the mutations in the residues 385-389 (E388 fixed) is shown in Table VII.
Table IV
PANNING OF LIBRARY (RESIDUES 308-314; H310 AND W313 FIXED) pCANTABSE-KUNKEL-muFcRn (MURINE FcRn) PANNING OUTPUT ENRICHMENT
+ FeRn - FcRn RATIO

1st Round 1.1 x 105 0.5 x 105 2 2nd Round 1 x 104 0.2 x 104 5 3rd Round 9 x 104 0.3 x 104 30 4th Round 3 x 105 2 x 104 15 Table V
PANNING OF LIBRARY (RESIDUES 251-256; 1253 FIXED) pCANTAB5E-KUNKEL-muFcRn PANNING OUTPUT ENRICHMENT
+ FcRn - FcRn RATIO

1st Round 2.5 x 105 1 x 105 2.5 2nd Round 6 x 104 2 x 104 3.0 3rd Round 8 x 105 4 x 104 20 4th Round 1.2 x 106 5 x 104 24 5th Round 3.0 x 106 6 x 104 50 Table VI
PANNING OF LIBRARY (RESIDUES 428-436;
H429, E430, A431, L432, AND H435 FIXED) pCANTAB5E-KUNKEL-muFcRn PANNING OUTPUT ENRICHMENT
+ FeRn - FcRn RATIO

1st Round 2.3 x 105 0.9 x 105 2.5 2nd Round 3 x 104 1 x 104 3 3rd Round 2 x 105 2 x 104 10 4th Round 8 x 105 5 x 104 16 Table VII
PANNING OF LIBRARY (RESIDUES 385-389; E388 FIXED) pCANTAB5E-KUNKEL-muFcRn PANNING OUTPUT ENRICHMENT
+ FcRn - FcRn RATIO
1st Round 4.2 x 105 3.8 x 105 1.1 2nd Round 5 x 104 0.3 x 104 17 3rd Round 3.5 x 105 1 x 104 35 4th Round 5.5 x 105 4 x 104 14 5th Round 7.5 x 105 5 x 104 15 6th Round 2 x 106 1 x 105 20 6.3 IDENTIFICATION OF ISOLATED CLONES FROM
PANNING

After each panning process, phage were isolated and the nucleic acids encoding the expressed peptides which bound to FeRn were sequenced by a standard sequencing method such as by dideoxynucleotide sequencing (Sanger et al., Proc. Natl.
Acad. Sci USA, 74:5463-5467, 1977) using a ABI3000 genomic analyzer (Applied Biosystems, Foster City, CA).
As a result of panning, two mutants were isolated from the phage library containing mutations in residues 308-314 (H310 and W313 fixed), thirteen mutants from the library for residues 251-256 (1253 fixed), six mutants from the library for residues 428-436 (H429, E430, A431, L432, and H435 fixed), and nine mutants from the library for residues 385-389 (E388 fixed). The mutants isolated from the libraries are listed in Table VIII.
Table VIII
MUTANTS ISOLATED BY PANNING
LIBRARY MUTANTS*

) 251-256 Leu Tyr Ile Thr Arg Glu (SEQ ID NO: 90 Leu Tyr Ile Ser Arg Thr (SEQ ID NO:91) Leu Tyr Ile Ser Arg Ser (SEQ ID NO:92) Leu Tyr Ile Ser ArgArg (SEQ ID NO:93) LIBRARY MUTANTS*
Leu Tyr Ile Ser Arg Gln (SEQ ID NO:94) Leu Trp Ile Ser Arg Thr (SEQ ID NO:95) Leu Tyr Ile Ser Leu Gln (SEQ ID NO:96) Leu Phe Ile Ser Arg Asp (SEQ ID NO:97) Leu Phe Ile Ser Arg Thr (SEQ ID NO:98) Leu Phe Ile Ser Arg Arg (SEQ ID NO:99) Leu Phe Ile Thr Gly Ala (SEQ ID NO: 100) Leu Ser Ile Ser Arg Glu (SEQ ID NO:101) Arg Thr Ile Ser Ile Ser (SEQ ID NO:102) 308-314 Thr Pro His Ser Asp TM Leu (SEQ ID NO:103) Ile Pro His Glu Asp Trp Leu (SEQ ID NO:104) 385-389 Arg Thr Arg Glu Pro (SEQ ID NO:105) Asp Pro Pro Glu Ser (SEQ ID NO:106) Ser Asp Pro Glu Pro (SEQ ID NO: 107) Thr Ser His Glu Asn (SEQ ID NO:108) Ser Lys Ser Glu Asn (SEQ ID NO:109) His Arg Ser Glu Asn (SEQ ID NO:110) Lys Ile Arg Glu Asn (SEQ ID NO: 111) Gly Ile Thr Glu Ser (SEQ ID NO: 112) Ser Met Ala Glu Pro (SEQ ID NO: 113) 428-436 Met His Glu Ala Leu Arg Tyr His His (SEQ ID NO:114) Met His Glu Ala Leu His Phe His His (SEQ ID NO: 115) Met His Glu Ala Leu Lys Phe His His (SEQ ID NO: 116) Met His Glu Ala Leu Ser Tyr His Arg (SEQ ID NO:117) Thr His Glu Ala Leu His Tyr His Thr (SEQ ID NO: 118) Met His Glu Ala Leu His Tyr His Tyr (SEQ ID NO: 119) * Substituting residues are indicated in bold face The underlined sequences in Table VIII correspond to sequences that occurred 10 to 20 times in the final round of panning and the sequences in italics correspond to sequences that occurred 2 to 5 times in the final round of panning. Those sequences that are neither underlined nor italicized occurred once in the final round of panning.

6.4 EXPRESSION AND PURIFICATION OF SOLUBLE
MUTANT HINGE-FC REGION
The genes encoding mutated hinge-Fc fragments are excised with appropriate restriction enzymes and recloned into an expression vector, for example, VopelBhis (Ward, J. Mol. Biol., 224:885-890, 1992). Vectors containing any other type of tag sequence, such as c-myc tag, decapeptide tag (Huse et al., Science, 246:1275-1281, 1989), Flag' (Immunex) tags, can be used. Recombinant clones, such as E. coli, are grown and induced to express soluble hinge-Fc fragments, which can be isolated from the culture media or cell lysate after osmotic shock, based on the tag used, or by any other purification methods well known to those skilled in the art and characterized by the methods as listed below.
6.5 CONSTRUCTION, PRODUCTION AND
PURIFICATION OF IgG1 VARIANTS

Representative Fc mutations such as 1253A, M252Y/S254T/T256E, M252W, M252Y, M252Y/T256Q, M252F/T256D, V308T/L309P/Q311S, G385D/Q386P/N389S, G385R/Q386T/P387R/N389P, H433K/N434F/Y436H, and N434F/Y436 were incorporated into the human IgGl MEDI-493 (SYNAGIS ) (Johnson et al., 1997, supra). The heavy chain was subjected to site-directed mutagenesis using a Quick Change Mutagenesis kit (Stratagene, La Jolla, CA) according to the manufacturer's instructions and sequences were verified by didoxynucleotide sequencing using a ABI3000 (Applied Biosystems, Foster City, CA) sequencer. The different constructions were expressed transiently in human embryonic kidney 293 cells using a CMV immediate-early promoter and dicistronic operon in which IgGl/VH is cosecreted with IgGl/VL (Johnson et al., 1997, supra). Transfection was carried out using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) and standard protocols. IgGs were purified from the conditioned media directly on 1 ml HiTrap protein A columns according to the manufacterer's instructions (APBiotech).

6.6 CHARACTERIZATION OF MUTATED HINGE-FC REGION
6.6.1 IN VITRO CHARACTERIZATION HPLC AND
SDS-PAGE
Following the purification, general characteristics such as molecular weight and bonding characteristics of the modified hinge-Fe fragments may be studied by various methods well known to those skilled in the art, including SDS-PAGE and HPLC.

FcRn binding assay Binding activity of modified hinge-Fc fragments can be measured by incubating radio-labeled wild-type hinge-Fe or modified hinge-Fc with the cells expressing either mouse or human FeRn. Typically, endothelial cell lines such as SV40 transformed endothelial cells (SVEC) (Kim et al., J Immunol., 40:457-465, 1994) are used.
After incubation with the hinge-Fc fragments at 37 C for 16-18 hours, the cells are washed with medium and then detached by incubation with 5 mM Na2EDTA in 50 mM phosphate buffer, pH 7.5, for 5 minutes. The radioactivity per 10' cells is measured.
Then, the cells are resuspended in 2 ml of 2.5 mg/ml CHAPS, 0.1 M Tris-HC1 pH 8.0 containing 0.3 mg/ml PMSF, 25 mg/ml pepstatin and 0.1 mg/ml aprotinin and incubated for 30 minutes at room temperature. The cell suspension is then centrifuged and the supernatant separated. The radioactivity of the supernatant is measured and used to calculate the amount of the hinge-Fc fragments extracted per 10' cells.
The Kd for the interaction of wild type human IgG1 with inurine and human FeRn (269 and 2527 nM, respectively) agree well with the values determined by others (265 and 2350 nM, respectively, Firan et al., 2001, supra). The 1253A mutation virtually abolishes binding to human and murine FcRn, as reported by others (Kim et al., Eur. I
Immunol., 29:2819-2825, 1991; Shields et al., J. Biol. Chem., 276:6591-6604, 2001). This is not the result of misfolding of the antibody as this mutant retains the same specific activity than the wild type molecule (SYNAGIS ) in a microneutralization assay (Johnson et al., 1997, supra; data not shown).
Human IgG1 mutants with increased binding affinity towards both murine and human FcRn were generated (Table VIII). Improvements in complex stability were overall less marked for the human IgG1-human FeRn pair than for the human IgGl-murine FeRn compared to wild type IgG1 were 30-(AAG = 2.0 kcal/mol for N434F/Y436H) and 11-(AAG
= 1.4 kcal/mol for M252Y/S254Y/S254T/T256E) fold, respectively. However, ranking of the most critical positions remain unchanged when comparing human and murine FcRn: the largest increases in IgG1-murine FcRn complex stability (AAG > 1.3 kcal/mol) occurred on mutations at positions 252, 254, 256 (M252Y/S254T/T256E and M252W) and 433, 434, 436 (H433K/N434F/Y436H and N434F/Y436H). Likewise, the same mutations were found to have the most profound impact on the IgGI-human FeRn interaction and also resulted in the largest increases in complex stability (AiG > 1.0 kcal/mol). Substitutions at positions 308, 309, 311, 385, 386, 387 and 389 had little or no effect on the stability of the complexes involving human or murine FcRn (AAG < 0.5 kcal/mol). Residues at the center of the Fc-FcRn combining site contribute significantly more to improvement in complex stability than residues at the periphery (FIG. 9).
Efficient binding of human Fc to murine FcRn apparently requires the presence of several wild type Fc residues. For example, leucine is very conserved at 251, arginine at 255, aspartic acid at 310, leucine at 314 and methionine at 428 (FIG. 6). Another specificity trend is observed when one considers positions 308, 309, and 311 where threonine, proline, and serine, respectively, are very strongly favored over the corresponding wild type residues (FIG. 6). However, generation of this strong consensus sequences does not correlate with the magnitude of increase in affinity as V308T/L309P/Q31 1S
binds less than 2-fold better than the wild type IgGI to both human and murine FcRn (Table IX).
Increases in affinity can be strongly dependent upon residue substitution at one `hot spot' position. For example, the single mutation M252Y causes an increase in binding to murine FcRn by 9-fold, whereas additional mutations bring little (M252Y/S254T/T256E) or no (M252Y/T256Q) added benefit. The same trend is observed for the human receptor, although to a lesser extent. Indeed, M252Y/S254T/T256E
shows a marked improvement of 2.5-fold in affinity compared to M252Y. This probably reflects the differences between the binding site of human and murine FcRn (West and Bjorkman, Biochemistry, 39:9698-9708, 2000).
Phage-derived IgGI mutants exhibiting a significant increase in affinity towards murine FcRn (AAG > 1.3 kcal/mol) also showed significant binding activity to the receptor at pH 7.2 when compared to wild type IgGI (FIGs. 8A-H). IgGI mutants with moderate increase in affinity (OMG < 0.3 kcal/mol) bound very poorly at pH 7.2 (data not shown). In contrast, IgGI mutants with large (BOG > 1.0 kcal/mol) increase in affinity towards human FcRn exhibited only minimal binding at pH 7.4 when compared to wild type IgGI (FIGs. 8A-H).

Table IX
DISSOCIATION CONSTANTS AND RELATIVE FREE ENERGY
CHANGES FOR THE BINDING OF IgG1/FC MUTANTS TO
MURINE AND HUMAN FcRn*
MUTANT Dissociation AAG Dissociation AAG
Constant (kcal/mol) Constant (kcal/mol) Fc/Murine Fc/Human FcRn (nM) FcRn (mM) wild type 269 1 2527 117 M252Y/S254T/T256E 27 6 1.4 225 10 1.4 M252W 30+1 1.3 408 24 1.1 M252Y 41 7 1.1 532 37 0.9 M252Y/T256Q 39 8 1.1 560 102 0.9 M252F/T256D 52 9 1.0 933 170 0.6 V308T/L309P/Q311S 153 23 0.3 1964 84 0.1 G385D/Q386P/N389S 187 10 0.2 2164 331 0.1 G385R/Q386T/P387R/N389P 147 24 0.4 1620 61 0.3 H433K/N434F/Y436H 14 2 1.8 399 47 1.1 N434F/Y436H 9 1 2.0 493 7 1.0 *Affinity measurements were carried out by BlAcore as described above. Residue numbering is according to EU (Kabat et al., 1991, supra). Differences in free energy changes are calculated as the differences between the Ags of wild type and mutant reactions (AAG = AG
wild type - AG mutant). NB, no binding. NA, not-applicable.

FeRn-mediated transfer assay This assay follows the protocol disclosed in PCT publication WO 97/34631.
Radiolabeled modified hinge-Fc fragments at various concentration (1 g/ml-1 mg/ml) are added to the one side of the transwell and the transfer of the fragments mediated by FcRn-expressing monolayer of the cells can be quantitated by measuring the radioactivity on the other side of the transwell.

6.6.2 IN VIVO PHARMACOKINETIC STUDY
In order to determine the half-life of the modified IgG hinge-Fe, modified hinge-Fc fragments are radiolabelled with 1211 (approximate specific activity of 10' cpm/ g) and dissolved in saline (pH 7.2). The solution is injected intravenously into BALB/c mice (Harlan, Indianapolis, IN), which have been given NaI-containing water previously to block the thyroid, in a volume not more than 150 l and with a radioactivity of 10 x 106-50 x 106 cpm. The mice are bled from the retro-orbital sinus at various time points, for example, at 3 minutes to 72 hours after the injection, into heparinized capillary tubes and the plasma collected from each sample is counted for radioactivity.
To generate the data provided in FIG. 10, 10 animals were used for each molecule assayed with 2.5 g of antibody injected per animal. Antibody serum levels were determined using an anti-human IgG ELISA (FIG. 10). There seems to be an inverse correlation between affinity to mouse FcRn and persistence in serum. This might be due to the significant amount of binding of the mutants observed at pH 7.2, which leads to the sequestration (i.e., lack of release in the serum) of the molecules.
Preliminary data (not shown) suggests increased transport of the mutants to the lung. Additionally, since the mutants exhibit lower levels of binding to human FcRn than murine FcRn (see FIGS. 8A-H), antibody sermn levels are expected to be higher in primates and humans.
6.6.3 SURFACE PLASMON RESONANCE ANALYSES
The interaction of soluble murine and human FcRn with immobilized human IgG1 variants was monitored by surface plasmon resonance detection using a BlAcore 3000 instrument (Pharmacia Biosensor, Uppsala, Sweden). No aggregated material which could interfere with affinity measurements (van der Merwe et al., EMBO J., 12:4945-4954, 1993;
van der Merwe et al., Biochemistry, 33:10149-10160, 1994) was detected by gel filtration.
Protein concentrations were calculated by the bicinchoninic acid (BCA) method for both human and murine FcRn or using the 1 % extinction coefficient at 280 nm of 1.5 for IgG1 wild type and variants. The latter were coupled to the dextran matrix of a CM5 sensor chip (Pharmacia Biosensor) using an Amine Coupling Kit as described (Johnson et al., supra).
The protein concentrations ranged from 3-5 g/ml in 10 mM sodium acetate, pH
5Ø The activation period was set for 7 minutes at a flow rate of 10 l/min and the immobilization period was set to between 10 and 20 minutes at a flow rate of 10 1/min.
Excess reactive esters were quenched by injection of 70 pl of 1.0 methanolamine hydrochloride, pH 8.5.
This typically resulted in the immobilization of between 500 and 4000 resonance units (RU).
Human and murine FcRn were buffer exchanged against 50 mM PBS buffer pH 6.0 containing 0.05% Tween 20. Dilutions were made in the same buffer. All binding experiments were performed at 25 C with concentrations ranging from 120 to 1 g/ml at a flow rate of 5 to 10 l/min; data were collected for 25 to 50 minutes and three 1-minute pulses of PBS buffer pH 7.2 were used to regenerate the surfaces. FcRn was also flowed over an uncoated cell and the sensorgrams from these blank runs subtracted from those obtained with IgG1-coupled chips. Runs were analyzed using the software BlAevaluation 3.1 (Pharmacia). Association constants (KAs) were determined from Scatchard analysis by measuring the concentration of free reactants and complex at equilibrium after correction for nonspecific binding. In equilibrium binding BlAcore experiments (Karlsson et al., 1991, supra; van der Merwe et al., 1993, supra; van der Merwe et al., 1994, supra;
Raghavan et al., Immunity, 1:303-315, 1994; Malchiodi et al., J. Exp. Med., 182:1833-1845, 1995), the concentration of the complex can be assessed directly as the steady-state response. The concentration of free analyte (human or murine FcRn) is equal to the bulk analyte concentration since analyte is constantly replenished during sample injection.
The concentration of free ligand on the surface of the sensor chip can be derived from the concentration of the complex and from the total binding capacity of the surface as KA =
Req/C(Rn,ax - Req) where C is the free analyte concentration, Reg is the steady-state response, and R. is the total surface binding capacity. Rearranging, the equation reads:
Req/C = KA
R.. - KA Req=
A plot of Req/C versus Req at different analyte concentrations thus gives a straight line from which KA can be calculated (see Table IX). Errors were estimated as the standard deviation for two or three independent determinations and were <20%.
Representative mutations identified after panning libraries 1 through 4 (FIG. 6, Table VIII) were introduced into the Fc portion of a human IgGl.
Injection of different concentrations of human or murine FcRn over the immobilized IgGi variants gave concentration-dependent binding. Typical resonance profiles for equilibrium binding of the mutant M252Y/S254T/T256E to murine and human FcRn are shown in FIGs. 7A and B.
To estimate apparent KAs, concentrations of FcRn ranging from 120 to 1 g/ml were used. In all cases, equilibrium (or near-equilibrium) binding levels were reached within 50 minutes. To estimate the increase in RU resulting from the non specific effect of protein on the bulk refractive index, binding of FcRn to an uncoated cell was measured and the sensorgrams from these blank runs subtracted from those obtained with IgGl-coupled chips.
The scatchard plots for the binding of the mutant M252Y/S254T/T256E to murine and human FcRn are shown in FIGs. 7C and D. The plots were all linear, and apparent KAs were calculated from the relevant slopes. Measurements were carried out in duplicate or triplicate and confirmed that the immobilized IgGs retained their original binding activity.
Since there are two non-equivalent binding sites on mouse IgGI for murine FcRn with affinities of < 130 nM and 6 M (Sanchez et al., Biochemistry, 38:9471-9476, 1999; Schuck et al, Mol. Immunol., 36:1117-1125, 1999; Ghetie and Ward, Ann.
Rev.
Immunol., 18:739-766, 2000), the receptor was used in solution to avoid avidity effects that arise when IgG i binds to immobilized FcRn. Consistent with this, systematically higher affinities are observed when FcRn, rather than IgG, immobilized on the biosensor chip (Popov et al., 1996, supra; Vaughn and Bjorkman, Biochemistry, 36:9374-9380, 1997;
Martin and Bjorkman, Biochemistry, 38:12639-12647; West and Bjorkman, Biochemistry, 39:9698-9708, 2000). Under our experimental BlAcore conditions, mainly interactions corresponding to the higher-affinity association (i.e. single liganded-recptor) are measured, according for the linearity of the scatchard plots (FIGs. 7C and D).
BlAcore analysis was also used to compare the affinity of wild type IgGI and IgGI mutants. Phage-derived IgGI mutants exhibiting a significant increase in affinity towards murine FcRn at pH 6.0 (MG a 1.0 kcal/mol) also shoed significant binding to the mouse receptor at pH 7.2 with SPR signalpH7.4/SPR signalpH6.a > 0.6 at saturation. IgGI
mutants with moderate increase in affinity towards murine FcRn at pH 6.0 (MMG
< 0.4 kcal/mol) bound very poorly to the mouse receptor at pH 7.2. In contrast, IgGI
mutants exhibiting large affinity increase towards human FcRn at pH 6.0 (MG z 1.0 kcal/mol) only showed minimal binding to the human receptor at pH 7.4 with SPR
signalLH7.4/SPR signalpH6.0 < 0.15 at saturation.
Those skilled in the art will recognize, or be able to ascertain using no more routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (86)

The embodiments of the present invention for which an exclusive property or privilege is claimed are defined as follows:
1. A modified molecule comprising a protein or non-protein agent and an IgG constant domain, wherein the IgG constant domain comprises a human CH2 domain in which there is an amino acid substitution at amino acid residue 252, numbered according to the EU index of Kabat, wherein the modified molecule has an increased half-life compared to the half-life of a molecule comprising the protein or the non-protein agent and a corresponding IgG constant domain comprising a human CH2 wild-type domain, and wherein the substitution at amino acid residue 252 is a substitution with tyrosine, phenylalanine, tryptophan or threonine.
2. The modified molecule according to claim 1, wherein the IgG constant domain is a human IgG constant domain.
3. The modified molecule according to claim 2, wherein the human IgG
constant domain is IgG1, IgG2, IgG3 or IgG4.
4. The modified molecule according to claim 3, wherein the human IgG
constant domain is IgG1.
5. The modified molecule according to any one of claims 2 to 4, wherein the IgG constant domain of the modified molecule comprises one or more amino acid substitutions at one or more of amino acid residues 251, 253-256, 285-290, 308-314, 385-389, and 428-436 relative to a wild-type human IgG constant domain.
6. The modified molecule according to any one of claims 2 to 4, wherein the IgG constant domain of the modified molecule comprises one or more amino acid substitutions at one or more of amino acid residues 254, 256, 309, 311, 314, 428, 433, 434 or 436 relative to a wild-type human IgG constant domain.
7. The modified molecule according to any one of claims 1 to 4, wherein the IgG constant domain of the modified molecule comprises an amino acid substitution at amino acid residue 254 and an amino acid substitution at amino acid residue 256 relative to a wild-type human CH2 domain.
8. The modified molecule according to claim 6 or 7, wherein the IgG
constant domain of the modified molecule comprises one or more amino acid substitutions at one or more of amino acid residues 385, 386, 387 or 389 relative to a wild-type human IgG
constant domain.
9. The modified molecule according to claim 5 or 6, wherein an amino acid substitution at amino acid residue 254 is a substitution with threonine;
the amino acid substitution at amino acid residue 256 is a substitution with phenylalanine, serine, arginine, glutamine, glutamic acid, aspartic acid, alanine or asparagine; the amino acid substitution at amino acid residue 309 is a substitution with proline; the amino acid substitution at amino acid residue 311 is a substitution with serine, glutamic acid or leucine; the amino acid substitution at amino acid residue 433 is a substitution with lysine, arginine, serine, isoleucine, proline, or glutamine; and the amino acid substitution at amino acid residue 434 is a substitution with histidine, phenylalanine or tyrosine.
10. The modified molecule according to claim 5 or 6, wherein the amino acid substitution at amino acid residue 252 is a substitution with tyrosine;
the amino acid substitution at amino acid residue 254 is a substitution with threonine; the amino acid substitution at amino acid residue 256 is glutamic acid; the amino acid substitution at amino acid residue 433 is a substitution with lysine; the amino acid substitution at amino acid residue 434 is a substitution with phenylalanine; and the amino acid substitution at amino acid residue 436 is a substitution with histidine.
11. The modified molecule according to claim 7, wherein the amino acid substitution at amino acid residue 254 is a substitution with threonine and the amino acid substitution at amino acid residue 256 is a substitution with phenylalanine, serine, arginine, glutamine, glutamic acid, aspartic acid, alanine or asparagine.
12. The modified molecule according to claim 7, wherein the amino acid substitution at amino acid residue 252 is a substitution with tyrosine; the amino acid substitution at amino acid residue 254 is a substitution with threonine; and the amino acid substitution at amino acid residue 256 is a substitution with glutamic acid.
13. The modified molecule according to claim 7, wherein the amino acid substitution at amino acid residue 252 is a substitution with tyrosine; the amino acid substitution at amino acid residue 254 is a substitution with threonine; and the amino acid substitution at amino acid residue 256 is a substitution with phenylalanine, serine, arginine, glutamine, glutamic acid, aspartic acid, alanine or asparagine.
14. The modified molecule according to claim 7, wherein the amino acid substitution at amino acid residue 254 is a substitution with threonine, and the amino acid substitution at amino acid residue 256 is a substitution with glutamic acid.
15. The modified molecule according to claim 8, wherein the amino acid substitution at amino acid residue 385 is a substitution with arginine, aspartic acid, serine, threonine, histidine, lysine, alanine or glycine; the amino acid substitution at amino acid residue 386 is a substitution with threonine, proline, aspartic acid, serine, lysine, arginine, isoleucine or methionine; the amino acid substitution at amino acid residue 387 is a substitution with arginine, histidine, serine, threonine or alanine; and the amino acid substitution at amino acid residue 389 is a substitution with proline, serine or asparagine.
16. The modified molecule according to claim 5 or 6, wherein the amino acid substitution at amino acid residue 428 is a substitution with methionine, threonine, leucine, phenylalanine or serine.
17. The modified molecule according to claim 5 or 6, wherein the amino acid substitution at amino acid residue 314 is a substitution with alanine.
18. The modified molecule according to claim 5, wherein the amino acid residue substitution at amino acid residue 251 is a substitution with arginine; the amino acid substitution at amino acid residue 254 is a substitution with threonine; the amino acid substitution at amino acid residue 255 is a substitution with leucine, glycine or isoleucine; the amino acid substitution at amino acid residue 256 is a substitution with phenylalanine, serine, arginine, glutamine, glutamic acid, aspartic acid, alanine or asparagine; the amino acid substitution at amino acid residue 308 is a substitution with a threonine or isoleucine; the amino acid substitution at amino acid residue 309 is a substitution with proline; the amino acid substitution at amino acid residue 311 is a substitution with serine, glutamic acid or leucine; the amino acid substitution at amino acid residue 312 is a substitution with alanine;
the amino acid substitution at amino acid residue 314 is a substitution with alanine; the amino acid substitution at amino acid residue 385 is a substitution with arginine, aspartic acid, serine, threonine, histidine, lysine, alanine or glycine; the amino acid substitution at amino acid residue 386 is a substitution with threonine, proline, aspartic acid, serine, lysine, arginine, isoleucine or methionine; the amino acid substitution at amino acid residue 387 is a substitution with arginine, histidine, serine, threonine, or alanine; the amino acid substitution at amino acid residue 389 is a substitution with proline, asparagine or serine; the amino acid substitution at amino acid residue 428 is a substitution with methionine, threonine, leucine, phenylalanine or serine; the amino acid substitution at amino acid residue 433 is a substitution with lysine, arginine, serine, isoleucine, proline, glutamine or histidine; the amino acid substitution at amino acid residue 434 is a substitution with phenylalanine, tyrosine or histidine; and the amino acid substitution at amino acid residue 436 is a substitution with histidine, asparagine, arginine, threonine, lysine, or methionine.
19. The modified molecule according to any one of claims 2 to 18, wherein the IgG constant domain of the modified molecule has a higher affinity for FcRn than a wild-type human IgG constant domain.
20. The modified molecule according to claim 19, wherein the IgG
constant domain of the modified molecule has a higher affinity for FcRn than a wild-type human IgG constant domain at pH 6.0 than at pH 7.4.
21. The modified molecule according to claim 2 to 20, wherein the modified molecule is a modified IgG.
22. The modified molecule according to claim 21, wherein the modified IgG is a modified human or humanized IgG.
23. The modified molecule according to claim 1 to 20, wherein the modified molecule comprises a protein agent.
24. The modified molecule according to claim 1 to 20, wherein the modified molecule comprises a non-protein agent.
25. The modified molecule according to any one of claims 1 to 20 or 23, wherein the modified molecule is a fusion protein comprising a non-IgG protein covalently linked to the IgG constant domain.
26. The modified molecule according to claim 24, wherein the non-protein agent is conjugated to the IgG constant domain.
27. The modified molecule according to any one of claims 21 or 22, wherein the modified IgG immunospecifically binds to a respiratory syncytial virus (RSV) antigen.
28. The modified molecule according to claim 27, wherein the RSV
antigen is a RSV F antigen.
29. The modified molecule according to claim 28, wherein the modified IgG comprises:
a. a variable heavy (VH) domain comprising the amino acid sequence of SEQ ID NO: 17 and a variable light (VL) domain comprising the amino acid sequence of SEQ ID NO:21, b. a VH domain comprising the amino acid sequence of SEQ ID NO:24 and a VL domain comprising the amino acid sequence of SEQ ID
NO:26, c. a VH domain comprising the amino acid sequence of SEQ ID NO:28 and a VL domain comprising the amino acid sequence of SEQ ID
NO:30, d. a VH domain comprising the amino acid sequence of SEQ ID NOS:33 and a VL domain comprising the amino acid sequence of SEQ ID
NO:34, e. a VH domain comprising the amino acid sequence of SEQ ID NO:36 and a VL domain comprising the amino acid sequence of SEQ ID
NO:38, f. a VH domain comprising the amino acid sequence of SEQ ID NO:40 and a VL domain comprising the amino acid sequence of SEQ ID
NO:42, g. a VH domain comprising the amino acid sequence of SEQ ID NO:44 and a VL domain comprising the amino acid sequence of SEQ ID
NO:46 h. a VH domain comprising the amino acid sequence of SEQ ID NO:51 and a VL domain comprising the amino acid sequence of SEQ ID
NO:52, i. a VH domain comprising the amino acid sequence of SEQ ID NO 7 and a VL domain comprising the amino acid sequence of SEQ ID
NO:54, j. a VH domain comprising the amino acid sequence of SEQ ID NO:55 and a VL domain comprising the amino acid sequence of SEQ ID
NO:56, k. a VH domain comprising the amino acid sequence of SEQ ID NO:55 and a VL domain comprising the amino acid sequence of SEQ ID
NO:124, 1. a VH domain comprising the amino acid sequence of SEQ ID NO:55 and a VL domain comprising the amino acid sequence of SEQ ID
NO:58, M. a VH domain comprising the amino acid sequence of SEQ ID NO:78 and a VL domain comprising the amino acid sequence of SEQ ID
NO:56, n. a VH domain comprising the amino acid sequence of SEQ ID NO:9 and a VL domain comprising the amino acid sequence of SEQ ID
NO:60, o. a VH domain comprising the amino acid sequence of SEQ ID NO:78 and a VL domain comprising the amino acid sequence of SEQ ID
NO:62, p. a VH domain comprising the amino acid sequence of SEQ ID NO:78 and a VL domain comprising the amino acid sequence of SEQ ID
NO:64, q. a VH domain comprising the amino acid sequence of SEQ ID NO:78 and a VL domain comprising the amino acid sequence of SEQ ID
NO:65, r. a VH domain comprising the amino acid sequence of SEQ ID NO:67 and a VL domain comprising the amino acid sequence of SEQ ID
NO:68, s. a VH domain comprising the amino acid sequence of SEQ ID NO:70 and a VL domain comprising the amino acid of SEQ ID NO:71, or t. a VH domain comprising the amino acid sequence of SEQ ID NO:48 and a VL domain comprising the amino acid sequence of SEQ ID
NO:74.
30. The modified molecule according to claim 28, wherein the modified IgG comprises:
a. a VH complementarily determining region (CDR) 1, a VH CDR2, a VH
CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:18, 19, 20, 22, 23 and 6, respectively, b. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:18, 25, 20, 22, 27 and 6, respectively, c. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:18, 25, 29, 31, 32 and 6, respectively, d. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 25, 29, 22, 35 and 6, respectively, e. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 37, 20, 39, 35 and 6, respectively, f. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS: 10, 41, 20, 22, 43 and 6, respectively, g. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 45, 20, 47, 43 and 6, respectively, h. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 45, 29, 31, 53 and 6, respectively, i. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:1, 2, 3, 14, 5 and 6, respectively, j. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 29, 14, 15 and 6, respectively, k. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 29, 14, 57 and 6, respectively, l. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 29, 14, 59 and 6, respectively, M. a VH CDR1, a VH CDR2, a VH CDR3, a, VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 79, 14, 15 and 6, respectively, n. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 12, 14, 15 and 61, respectively, o. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 79, 14, 63, and 6, respectively, p. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 79, 39, 15 and 6, respectively, q. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 2, 79, 14, 66 and 6, respectively, r. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 19, 29, 31, 69 and 6, respectively, s. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 25, 29, 72, 73 and 6, respectively, or t. a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL CDR3 comprising the amino acid sequence of SEQ ID NOS:10, 19, 20, 39, 75 and 6, respectively.
31. The modified molecule according to claim 28, wherein the modified IgG comprises the VH domain and VL domain of palivizumab.
32. The modified molecule according to claim 28, wherein the modified IgG comprises the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2 and VL CDR3 of palivizumab.
33. The modified molecule according to claim 28, wherein the modified IgG comprises a VH domain comprising the amino acid sequence of SEQ ID NO:48 and a VL domain comprising the amino acid sequence of SEQ ID NO: 11.
34. The modified molecule according to claim 28, wherein the modified IgG comprises a VH CDR1, aVH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL
CDR3 comprising the amino acid sequence SEQ ID NOS.: 10, 19, 20, 39, 5, and 6, respectively.
35. The modified molecule according to claim 28, wherein the modified IgG comprises a VH domain comprising the amino acid sequence of SEQ ID NO:48 and a VL domain comprising the amino acid sequence of SEQ ID NO:49.
36. The modified molecule according to claim 28, wherein the modified IgG comprises a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL
CDR3 comprising the amino acid sequence of SEQ ID NOS::10, 19, 20, 39, 50, and 6, respectively.
37. The modified molecule according to claim 28, wherein the modified IgG comprises the a VH domain comprising the amino acid sequence of SEQ ID
NO:48 and a VL domain comprising the amino acid sequence of SEQ ID NO:76.
38. The modified molecule according to claim 28, wherein the modified IgG comprises a VH CDR1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2 and a VL
CDR3 of comprising the amino acid sequence of SEQ ID NOS.:10, 19, 20, 39, 77, and 6, respectively.
39. The modified molecule according to claim 21 or 22, wherein the modified IgG immunospecifically binds to HER2, tumor necrosis factor-alpha (TNF-.alpha.), transforming growth factor-.beta. (TGF-.beta.), interleukin-4 (IL-4), interleukin-5 (IL-5), interleukin-8(IL-8), CD2, CD3, CD4, CD11a, CD14, CD18, CD20, CD22, CD23, CD25, CD33, CD52, CD64, CD80, CD147, CD40 ligand (CD40L), vascular endothelial growth factor (VEGF), intracellular adhesion molecule-3 (ICAM-3), epithelial growth factor receptor (EGFR), .alpha.v.beta.3 integrin, .alpha.4.beta.7 integrin, human leukocyte antigen (HLA), complement factor 5(C5), immunoglobulin E (IgE), glycoprotein II b/III a receptor, CA125, 17-IA cell surface antigen, Factor VII, GD3 epitope, human immunodeficiency glycoprotein 120 (HIV gp1120), hepatitis B virus (HBV) or cytomeglovirus (CMV), VLA-4, or .beta.2-integrin.
40. A pharmaceutical composition comprising the modified molecule according to any one of claims 1 to 39, and a pharmaceutically acceptable carrier.
41. A kit comprising the modified molecule according to any one of claims 1 to 39, in a container, and instructions for use.
42. A nucleic acid comprising a nucleotide sequence encoding the constant domain of the modified molecule according to any one of claims 1 to 39.
43. The nucleic acid of claim 42, which is an isolated nucleic acid.
44. A vector comprising the nucleic acid according to claim 42 or 43.
45. A host cell comprising or expressing the nucleic acid according to claim 42 or 43.
46. The host cell of claim 45, which is a CHO, VERY, BHK, HeLa, COS, MDCK, 293, 3T3, W138, or NS0 cell.
47. The host cell of claim 45 or 46, which is an isolated host cell.
48. A nucleic acid comprising a nucleotide sequence encoding the modified molecule according to claim 29, 31, 33, 35 or 37.
49. The nucleic acid of claim 48, which is an isolated nucleic acid.
50. A vector comprising the nucleic acid according to claim 48 or 49.
51. A host cell comprising or expressing the nucleic acid according to claim 48 or 49.
52. The host cell of claim 51, which is a CHO, VERY, BHK, HeLa, COS, MDCK, 293, 3T3, W138, or NS0 cell.
53. The host cell of claim 51 or 52, which is an isolated host cell.
54. The modified molecule according to any one of claims 1 to 26 or 39, for use in preventing a disease or disorder in a subject.
55. The modified molecule according to any one of claims 1 to 26 or 39, for use in treating a disease or disorder in a subject.
56. The modified molecule according to any one of claims 27 to 38, for use in preventing a disease or disorder in a subject.
57. The modified molecule according to any one of claims 27 to 38, for use in treating a disease or disorder in a subject.
58. The modified molecule according to any one of claims 27 to 38, for use in preventing a RSV infection or a symptom thereof in a subject.
59. The modified molecule according to any one of claims 27 to 38, for use in treating a RSV infection or a symptom thereof in a subject.
60. The modified molecule according to any one of claims 54 to 59, wherein the subject is a human.
61. Use of the modified molecule according to any one of claims 1 to 26 or 39 in the manufacture of a medicament for preventing a disease or disorder in a subject.
62. Use of the modified molecule according to any one of claims 1 to 26 or 39 in the manufacture of a medicament for treating a disease or disorder in a subject.
63. Use of the modified molecule according to any one of claims 27 to 38 in the manufacture of a medicament for preventing a disease or disorder in a subject.
64. Use of the modified molecule according to any one of claims 27 to 38 in the manufacture of a medicament for treating a disease or disorder in a subject.
65. Use of the modified molecule according to any one of claims 27 to 38 in the manufacture of a medicament for preventing a RSV infection or a symptom thereof in a subject.
66. Use of the modified molecule according to any one of claims 27 to 38 in the manufacture of a medicament for treating a RSV infection or a symptom thereof in a subject.
67. The use according to any one of claims 61 to 66, wherein the subject is in a human.
68. The modified molecule according to any one of claims 1 to 26 or 39, for use in the diagnosis of a disease or disorder.
69. The modified molecule according to any one of claims 27 to 38, for use in the diagnosis of a RSV infection.
70. An antibody conjugate comprising the modified molecule according to any one of claims 1 to 26 or 39 and a detectable substance.
71. An antibody conjugate comprising the modified molecule according to any one of claims 27 to 38 and a detectable substance.
72. An antibody conjugate comprising the modified molecule according to any one of claims 1 to 26 or 39 and a therapeutic moiety.
73. An antibody conjugate comprising the modified molecule according to any one of claims 27 to 38 and a therapeutic moiety.
74. A pharmaceutical composition comprising the antibody conjugate according to any one of claims 70 to 73, and a pharmaceutically acceptable carrier.
75. A kit comprising the antibody conjugate according to any one of claims 70 to 73, and instructions for use.
76. The antibody conjugate according to claim 72 or 73, for use in preventing a disease or disorder in a subject.
77. The antibody conjugate according to claim 72 or 73, for use in treating a disease or disorder in a subject.
78. An antibody conjugate of claim 76 or 77, wherein the subject is a human.
79. The antibody conjugate of claim 73, for use in preventing a RSV
infection or a symptom thereof in a human subject.
80. The antibody conjugate of claim 73, for use in treating a RSV infection or a symptom thereof in a human subject.
81. Use of the antibody conjugate of claim 72 or 73 in the manufacture of a medicament for preventing a disease or disorder in a subject.
82. Use of the antibody conjugate of claim 72 or 73 in the manufacture of a medicament for treating a disease or disorder in a subject.
83. The use of claim 81 or 82, wherein the subject is a human.
84. Use of the antibody conjugate of claim 73 in the manufacture of a medicament for preventing a RSV infection or a symptom thereof in a human subject.
85. Use of the antibody conjugate of claim 73 in the manufacture of a medicament for treating a RSV infection or a symptom thereof in a human subject.
86. A method of producing the modified molecule of any one of claims 27 to 38, the method comprising (a) transforming host cells with a vector comprising a promoter operably linked to the nucleotide sequence encoding the constant domain of the modified molecule, and (b) culturing said host cells to produce the modified molecule.
CA2431600A 2000-12-12 2001-12-12 Molecules with extended half-lives, compositions and uses thereof Expired - Lifetime CA2431600C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2766160A CA2766160A1 (en) 2000-12-12 2001-12-12 Modified molecules with increased affinity for fcrn, compositions, and uses thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US25488400P 2000-12-12 2000-12-12
US60/254,884 2000-12-12
US28976001P 2001-05-09 2001-05-09
US60/289,760 2001-05-09
PCT/US2001/048432 WO2002060919A2 (en) 2000-12-12 2001-12-12 Molecules with extended half-lives, compositions and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CA2766160A Division CA2766160A1 (en) 2000-12-12 2001-12-12 Modified molecules with increased affinity for fcrn, compositions, and uses thereof

Publications (2)

Publication Number Publication Date
CA2431600A1 CA2431600A1 (en) 2002-08-08
CA2431600C true CA2431600C (en) 2012-04-17

Family

ID=26944296

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2431600A Expired - Lifetime CA2431600C (en) 2000-12-12 2001-12-12 Molecules with extended half-lives, compositions and uses thereof

Country Status (12)

Country Link
US (8) US7083784B2 (en)
EP (5) EP3569610A3 (en)
JP (7) JP4336498B2 (en)
AT (1) ATE489395T1 (en)
AU (1) AU2002248184C1 (en)
CA (1) CA2431600C (en)
CY (1) CY1111283T1 (en)
DE (1) DE60143544D1 (en)
DK (1) DK1355919T3 (en)
ES (2) ES2727425T3 (en)
PT (1) PT1355919E (en)
WO (1) WO2002060919A2 (en)

Families Citing this family (762)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
EP2364997A3 (en) * 1999-01-15 2012-07-04 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
ATE474854T1 (en) * 2000-01-27 2010-08-15 Medimmune Llc RSV NEUTRALIZING ANTIBODIES WITH VERY HIGH AFFINITY
EP2341075A1 (en) * 2000-03-01 2011-07-06 MedImmune, LLC Antibodies binding to the f protein of a respiratory syncytial virus (rsv)
US6855493B2 (en) 2000-11-28 2005-02-15 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US7179900B2 (en) * 2000-11-28 2007-02-20 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
ES2727425T3 (en) * 2000-12-12 2019-10-16 Medimmune Llc Molecules with prolonged half-lives, compositions and uses thereof
US7658921B2 (en) * 2000-12-12 2010-02-09 Medimmune, Llc Molecules with extended half-lives, compositions and uses thereof
US20040002587A1 (en) * 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US20070122406A1 (en) 2005-07-08 2007-05-31 Xencor, Inc. Optimized proteins that target Ep-CAM
US20080260731A1 (en) * 2002-03-01 2008-10-23 Bernett Matthew J Optimized antibodies that target cd19
US20080254027A1 (en) * 2002-03-01 2008-10-16 Bernett Matthew J Optimized CD5 antibodies and methods of using the same
US20100311954A1 (en) * 2002-03-01 2010-12-09 Xencor, Inc. Optimized Proteins that Target Ep-CAM
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US20090042291A1 (en) * 2002-03-01 2009-02-12 Xencor, Inc. Optimized Fc variants
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US20070148171A1 (en) * 2002-09-27 2007-06-28 Xencor, Inc. Optimized anti-CD30 antibodies
US7317091B2 (en) * 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
AU2003276832A1 (en) * 2002-05-10 2004-02-25 Medimmune, Llc EphA2 AGONISTIC MONOCLONAL ANTIBODIES AND METHODS OF USE THEREOF
US20050152899A1 (en) * 2002-05-10 2005-07-14 Kinch Michael S. EphA2 agonistic monoclonal antibodies and methods of use thereof
US20040028685A1 (en) * 2002-05-10 2004-02-12 Kinch Michael S. EphA2 monoclonal antibodies and methods of use thereof
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US7425618B2 (en) * 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
US7217797B2 (en) * 2002-10-15 2007-05-15 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7365168B2 (en) 2002-10-15 2008-04-29 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
AU2003286467B2 (en) * 2002-10-15 2009-10-01 Abbvie Biotherapeutics Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US7700100B2 (en) 2003-01-13 2010-04-20 Macrogenics, Inc. FcγRIIB fusion proteins and compositions thereof
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US20090010920A1 (en) 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
CN1798767B (en) * 2003-04-10 2011-02-16 晶面生物技术公司 Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
US20050059592A1 (en) * 2003-04-11 2005-03-17 Kiener Peter A. EphA2 and hyperproliferative cell disorders
KR20110094361A (en) 2003-04-11 2011-08-23 메디뮨 엘엘씨 Recombinant il-9 antibodies and uses thereof
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
DK1639014T3 (en) * 2003-06-13 2011-01-17 Biogen Idec Inc Aglycosyl anti-CD154 (CD40 ligand) antibodies and uses thereof
JP4178514B2 (en) * 2003-06-27 2008-11-12 東洋紡績株式会社 Antibody screening method
US20050106667A1 (en) 2003-08-01 2005-05-19 Genentech, Inc Binding polypeptides with restricted diversity sequences
CA2536408A1 (en) 2003-08-22 2005-03-03 Biogen Idec Ma Inc. Improved antibodies having altered effector function and methods for making the same
EP1660094A4 (en) 2003-08-26 2009-09-16 Univ Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of bacterial infections
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
CA2545539A1 (en) * 2003-10-15 2005-04-28 Pdl Biopharma, Inc. Alteration of fc-fusion protein serum half-lives by mutagenesis of positions 250, 314 and/or 428 of the heavy chain constant region of ig
EP2385069A3 (en) 2003-11-12 2012-05-30 Biogen Idec MA Inc. Neonatal Fc rReceptor (FcRn)- binding polypeptide variants, dimeric Fc binding proteins and methods related thereto
WO2005063815A2 (en) * 2003-11-12 2005-07-14 Biogen Idec Ma Inc. Fcϝ receptor-binding polypeptide variants and methods related thereto
EP1697520A2 (en) * 2003-12-22 2006-09-06 Xencor, Inc. Fc polypeptides with novel fc ligand binding sites
EP2053062A1 (en) * 2004-03-24 2009-04-29 Xencor, Inc. Immunoglobin variants outside the Fc region
AU2005249360B2 (en) 2004-04-12 2011-07-21 Medimmune, Llc Anti-IL-9 antibody formulations and uses thereof
BRPI0510883B8 (en) 2004-06-01 2021-05-25 Genentech Inc drug-antibody conjugate compound, pharmaceutical composition, method of manufacturing a drug-antibody conjugate compound, and uses of a formulation, a drug-antibody conjugate and a chemotherapeutic agent, and a combination
US20150010550A1 (en) 2004-07-15 2015-01-08 Xencor, Inc. OPTIMIZED Fc VARIANTS
KR20070047327A (en) * 2004-07-26 2007-05-04 비오겐 아이덱 엠에이 아이엔씨. Anti-cd154 antibodies
DK1776384T3 (en) 2004-08-04 2013-09-02 Mentrik Biotech Llc VARIANT Fc REGIONS
US7659374B2 (en) * 2004-08-16 2010-02-09 Medimmune, Llc Eph receptor Fc variants with enhanced antibody dependent cell-mediated cytotoxicity activity
EP1778728A2 (en) * 2004-08-19 2007-05-02 Genentech, Inc. Polypeptide variants with altered effector function
WO2006031994A2 (en) * 2004-09-14 2006-03-23 Xencor, Inc. Monomeric immunoglobulin fc domains
US20060121042A1 (en) 2004-10-27 2006-06-08 Medimmune, Inc. Modulation of antibody specificity by tailoring the affinity to cognate antigens
EP1812068A4 (en) * 2004-10-29 2010-06-09 Medimmune Inc Methods of preventing and treating rsv infections and related conditions
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
WO2006053301A2 (en) * 2004-11-12 2006-05-18 Xencor, Inc. Fc variants with altered binding to fcrn
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US20070135620A1 (en) * 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
CA2594356C (en) 2005-01-05 2018-07-17 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H. Synthetic immunoglobulin domains with binding properties engineered in regions of the molecule different from the complementarity determining regions
WO2006076594A2 (en) * 2005-01-12 2006-07-20 Xencor, Inc. Antibodies and fc fusion proteins with altered immunogenicity
ES2565543T3 (en) * 2005-01-24 2016-04-05 Board Of Regents, The University Of Texas System Fc fusion constructs to phosphatidylserine binding and its therapeutic use
AU2006214121B9 (en) 2005-02-15 2013-02-14 Duke University Anti-CD19 antibodies and uses in oncology
US8999943B2 (en) 2005-03-14 2015-04-07 Board Of Regents, The University Of Texas System Antigene oligomers inhibit transcription
KR101374454B1 (en) 2005-03-31 2014-03-17 추가이 세이야쿠 가부시키가이샤 Methods for producing polypeptides by regulating polypeptide association
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
US11254748B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
ES2707152T3 (en) 2005-04-15 2019-04-02 Macrogenics Inc Covalent diabodies and uses thereof
WO2006116260A2 (en) * 2005-04-26 2006-11-02 Medimmune, Inc. Modulation of antibody effector function by hinge domain engineering
CA2607281C (en) 2005-05-05 2023-10-03 Duke University Anti-cd19 antibody therapy for autoimmune disease
EP1891111A1 (en) 2005-05-06 2008-02-27 ZymoGenetics, Inc. Il-31 monoclonal antibodies and methods of use
EP1896503B1 (en) * 2005-05-31 2014-10-29 Board of Regents, The University of Texas System IgG1 ANTIBODIES WITH MUTATED Fc PORTION FOR INCREASED BINDING TO FcRn RECEPTOR AND USES TEHEREOF
WO2007005612A2 (en) * 2005-07-01 2007-01-11 Medimmune, Inc. An integrated approach for generating multidomain protein therapeutics
CA2618681C (en) 2005-08-10 2015-10-27 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
DK1931709T3 (en) 2005-10-03 2017-03-13 Xencor Inc FC VARIETIES WITH OPTIMIZED FC RECEPTOR BINDING PROPERTIES
AU2006302254B2 (en) 2005-10-06 2011-05-26 Xencor, Inc. Optimized anti-CD30 antibodies
KR20080073293A (en) 2005-10-14 2008-08-08 메디뮨 엘엘씨 Cell display of antibody libraries
ES2577292T3 (en) 2005-11-07 2016-07-14 Genentech, Inc. Binding polypeptides with diversified VH / VL hypervariable sequences and consensus
AU2006336624B2 (en) 2005-11-17 2010-11-25 Board Of Regents, The University Of Texas System Modulation of gene expression by oligomers targeted to chromosomal DNA
JP5525729B2 (en) * 2005-11-28 2014-06-18 ゲンマブ エー/エス Recombinant monovalent antibody and production method thereof
EP2567973B1 (en) 2005-11-28 2014-05-14 Zymogenetics, Inc. IL-21 antagonists
US8957187B2 (en) 2005-12-02 2015-02-17 Genentech, Inc. Binding polypeptides and uses thereof
JP2009525986A (en) * 2006-02-03 2009-07-16 メディミューン,エルエルシー Protein preparation
US8389688B2 (en) 2006-03-06 2013-03-05 Aeres Biomedical, Ltd. Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
EP2009101B1 (en) * 2006-03-31 2017-10-25 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
DK3056568T3 (en) 2006-03-31 2021-11-01 Chugai Pharmaceutical Co Ltd PROCEDURES FOR CONTROL OF THE BLOOD PHARMACOKINETICS OF ANTIBODIES
CA2652945C (en) 2006-05-30 2015-06-02 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
WO2010123874A1 (en) 2009-04-20 2010-10-28 Oxford Biotherapeutics Ltd. Antibodies specific to cadherin-17
SI2029173T1 (en) 2006-06-26 2016-12-30 Macrogenics, Inc. Fc riib-specific antibodies and methods of use thereof
CA2656224C (en) 2006-06-26 2018-01-09 Macrogenics, Inc. Combination of fc.gamma.riib antibodies and cd20-specific antibodies and methods of use thereof
AT503861B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING T-CELL RECEPTORS
AT503902B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING IMMUNE LOBULINS
AT503889B1 (en) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F MULTIVALENT IMMUNE LOBULINE
CL2007002225A1 (en) 2006-08-03 2008-04-18 Astrazeneca Ab SPECIFIC UNION AGENT FOR A RECEIVER OF THE GROWTH FACTOR DERIVED FROM PLATES (PDGFR-ALFA); NUCLEIC ACID MOLECULA THAT CODIFIES IT; VECTOR AND CELL GUESTS THAT UNDERSTAND IT; CONJUGADO UNDERSTANDING THE AGENT; AND USE OF THE AGENT OF A
AU2007348941B2 (en) 2006-08-03 2011-08-04 Medimmune Limited Antibodies directed to alphaVbeta6 and uses thereof
DK2511301T3 (en) 2006-08-04 2018-03-12 Medimmune Ltd HUMAN ANTIBODIES AGAINST ERBB 2
RS53263B (en) 2006-08-14 2014-08-29 Xencor Inc. Optimized antibodies that target cd19
BRPI0715660B8 (en) 2006-09-01 2021-05-25 Zymogenetics Inc isolated antibody that binds to human IL-31; and, use of a monoclonal antibody
US8323653B2 (en) 2006-09-08 2012-12-04 Medimmune, Llc Humanized anti-CD19 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
AU2007299843B2 (en) 2006-09-18 2012-03-08 Xencor, Inc Optimized antibodies that target HM1.24
EP2407548A1 (en) * 2006-10-16 2012-01-18 MedImmune, LLC Molecules with reduced half-lives, compositions and uses thereof
PT2502938E (en) 2006-10-27 2015-06-05 Genentech Inc Antibodies and immunoconjugates and uses therefor
CN101678100A (en) 2006-12-06 2010-03-24 米迪缪尼有限公司 methods of treating systemic lupus erythematosus
US8652466B2 (en) 2006-12-08 2014-02-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
TW200831538A (en) 2006-12-19 2008-08-01 Genentech Inc VEGF-specific antagonists for adjuvant and neoadjuvant therapy and the treatment of early stage tumors
PL3199180T3 (en) 2007-03-08 2022-08-08 Humanigen, Inc. Epha3 antibodies for the treatment of solid tumors
EP1980269A1 (en) * 2007-04-13 2008-10-15 Katholieke Universiteit Leuven Prevention of staphylococcus biofilm formation
EP2068925A4 (en) 2007-05-07 2011-08-31 Medimmune Llc Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
DK2176298T3 (en) 2007-05-30 2018-02-12 Xencor Inc Methods and compositions for inhibiting CD32B-expressing cells
EP1997830A1 (en) 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
US7580304B2 (en) * 2007-06-15 2009-08-25 United Memories, Inc. Multiple bus charge sharing
US8921279B2 (en) 2007-06-26 2014-12-30 F-Star Biotechnologische Forschungs—und Entwicklungsges. m.b.H Display of binding agents
WO2009006520A1 (en) * 2007-07-03 2009-01-08 Medimmune, Llc Hinge domain engineering
JP2010535032A (en) 2007-07-31 2010-11-18 メディミューン,エルエルシー Multispecific epitope binding proteins and uses thereof
PE20140196A1 (en) 2007-08-09 2014-03-19 Boehringer Ingelheim Int ANTI-CD37 ANTIBODIES
CN101772574B (en) * 2007-08-21 2016-01-06 莫佛塞斯公司 The method for disulfide formation of improvement
EP3789400A1 (en) 2007-09-26 2021-03-10 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
JP5334319B2 (en) 2007-09-26 2013-11-06 中外製薬株式会社 Method for modifying isoelectric point of antibody by amino acid substitution of CDR
WO2009061818A1 (en) 2007-11-05 2009-05-14 Medimmune, Llc Methods of treating scleroderma
DK2219672T3 (en) 2007-11-09 2016-05-17 Peregrine Pharmaceuticals Inc The anti-VEGF antibody compositions and methods
LT2796466T (en) 2007-12-07 2018-02-26 Zymogenetics, Inc. Humanized antibody molecules specific for IL-31
KR101615215B1 (en) 2007-12-07 2016-04-25 지모제넥틱스, 인코포레이티드 Anti-human il-21 monoclonal antibodies
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
US8092804B2 (en) 2007-12-21 2012-01-10 Medimmune Limited Binding members for interleukin-4 receptor alpha (IL-4Rα)-173
EP2245064B1 (en) 2007-12-21 2014-07-23 Medimmune Limited BINDING MEMBERS FOR INTERLEUKIN-4 RECEPTOR ALPHA (IL-4Ralpha)
SI2808343T1 (en) 2007-12-26 2019-10-30 Xencor Inc Fc variants with altered binding to FcRn
KR20100107501A (en) 2008-01-18 2010-10-05 메디뮨 엘엘씨 Cysteine engineered antibodies for site-specific conjugation
EP2080770A1 (en) * 2008-01-21 2009-07-22 MorphoSys AG Proteinaceous binding molecules comprising purification tags
BR122020023189B1 (en) 2008-02-08 2022-02-01 Astrazeneca Ab Use of a pharmaceutical composition comprising an antibody specific for ifnar1
CA2720365C (en) 2008-04-02 2019-01-15 Macrogenics, Inc. Bcr-complex-specific antibodies and methods of using same
CN102046195A (en) 2008-04-02 2011-05-04 宏观基因有限公司 HER2/neu-specific antibodies and methods of using same
CA2721052C (en) 2008-04-11 2023-02-21 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
EP2280997A2 (en) * 2008-04-18 2011-02-09 Xencor, Inc. Human equivalent monoclonal antibodies engineered from nonhuman variable regions
EP2113255A1 (en) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Cytotoxic immunoglobulin
TW201016233A (en) * 2008-07-15 2010-05-01 Genentech Inc Methods of treating autoimmune diseases using CD4 antibodies
CA2735900A1 (en) 2008-09-19 2010-03-25 Medimmune, Llc Antibodies directed to dll4 and uses thereof
TWI440469B (en) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
KR102100066B1 (en) * 2008-10-14 2020-04-10 제넨테크, 인크. Immunoglobulin variants and uses thereof
US8298533B2 (en) 2008-11-07 2012-10-30 Medimmune Limited Antibodies to IL-1R1
JP5933975B2 (en) 2008-11-12 2016-06-15 メディミューン,エルエルシー Antibody preparation
JP6041489B2 (en) 2008-11-22 2016-12-07 ジェネンテック, インコーポレイテッド Use of anti-VEGF antibodies in combination with chemotherapy for the treatment of breast cancer
US8775090B2 (en) * 2008-12-12 2014-07-08 Medimmune, Llc Crystals and structure of a human IgG Fc variant with enhanced FcRn binding
US20120114667A1 (en) 2008-12-23 2012-05-10 Medimmune Limited TARGETED BINDING AGENTS DIRECTED TO a5BETA1 AND USES THEREOF
CN102341411A (en) 2008-12-31 2012-02-01 比奥根艾迪克Ma公司 Anti-lymphotoxin antibodies
EP2391384A4 (en) * 2009-01-29 2012-12-26 Medimmune Llc Human anti-il-6 antibodies with extended in vivo half-life and their use in treatment of oncology, autoimmune diseases and inflammatory diseases
CN102405237A (en) 2009-03-06 2012-04-04 卡罗拜奥斯制药公司 Treatment of leukemias and chronic myeloproliferative diseases with antibodies to epha3
KR20110136825A (en) 2009-03-09 2011-12-21 바이오아트라, 엘엘씨 Mirac proteins
NZ600915A (en) * 2009-03-16 2013-09-27 Cephalon Australia Pty Ltd Humanised antibodies with anti-tumour activity
TWI544077B (en) 2009-03-19 2016-08-01 Chugai Pharmaceutical Co Ltd Antibody constant region change body
US9228017B2 (en) 2009-03-19 2016-01-05 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
EP2233500A1 (en) 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
EP2679600A1 (en) 2009-03-25 2014-01-01 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
DK2437767T3 (en) * 2009-06-01 2015-09-28 Medimmune Llc MOLECULES WITH EXTENDED half-lives and uses thereof
CN102802661B (en) 2009-06-22 2016-01-13 米迪缪尼有限公司 For the engineered Fc district of site-specific conjugation
PL2464664T3 (en) 2009-08-13 2016-02-29 Crucell Holland Bv Antibodies against human respiratory syncytial virus (rsv) and methods of use
EP2464661B1 (en) 2009-08-13 2018-01-17 The Johns Hopkins University Methods of modulating immune function with anti-b7-h7cr antibodies
WO2011022264A1 (en) 2009-08-15 2011-02-24 Genentech, Inc. Anti-angiogenesis therapy for the treatment of previously treated breast cancer
JP5813641B2 (en) 2009-08-24 2015-11-17 アムニクス オペレーティング インコーポレイテッド Coagulation factor IX composition and methods of making and using the same
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
IN2012DN03025A (en) 2009-09-09 2015-07-31 Ct Se Llc
US20110189183A1 (en) 2009-09-18 2011-08-04 Robert Anthony Williamson Antibodies against candida, collections thereof and methods of use
WO2011037158A1 (en) 2009-09-24 2011-03-31 中外製薬株式会社 Modified antibody constant regions
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
US20120231004A1 (en) 2009-10-13 2012-09-13 Oxford Biotherapeutic Ltd. Antibodies
JP2013508292A (en) 2009-10-14 2013-03-07 カロバイオス ファーマシューティカルズ インコーポレイティッド Antibodies against EphA3
US8435511B2 (en) 2009-10-22 2013-05-07 Genentech, Inc. Anti-hepsin antibodies and methods using same
PL2496691T3 (en) 2009-11-02 2017-09-29 University Of Washington Therapeutic nuclease compositions and methods
CA2780221A1 (en) 2009-11-04 2011-05-12 Fabrus Llc Methods for affinity maturation-based antibody optimization
US8932588B2 (en) 2009-11-05 2015-01-13 Teva Pharmaceuticals Australia Pty. Ltd. Treatment of cancer involving mutated KRAS or BRAF genes
KR101968766B1 (en) 2009-11-05 2019-04-12 제넨테크, 인크. Methods and composition for secretion of heterologous polypeptides
US8881354B2 (en) 2009-11-16 2014-11-11 Jtekt Corporation Tool radius adjusting system for boring holder, tool radius adjusting method in machine tool, and machine tool
RS56469B1 (en) 2009-11-24 2018-01-31 Medimmune Ltd Targeted binding agents against b7-h1
SI2516468T1 (en) 2009-12-23 2016-06-30 Synimmune Gmbh Anti-flt3 antibodies and methods of using the same
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
AU2011205316B2 (en) 2010-01-13 2015-05-28 Oncomed Pharmaceuticals, Inc. Notch1 binding agents and methods of use thereof
WO2011091078A2 (en) 2010-01-19 2011-07-28 Xencor, Inc. Antibody fc variants with enhanced complement activity
WO2011089211A1 (en) 2010-01-22 2011-07-28 Synimmune Gmbh Anti-cd133 antibodies and methods of using the same
RU2573994C2 (en) 2010-02-10 2016-01-27 Иммьюноджен, Инк Anti-cd20 antibodies and thereof application
WO2011102845A1 (en) * 2010-02-18 2011-08-25 Transtech Pharma, Inc. Rage fusion protein compositions and methods of use
EP3696194A1 (en) 2010-02-23 2020-08-19 F. Hoffmann-La Roche AG Anti-angiogenesis therapy for the treatment of ovarian cancer
TWI448539B (en) 2010-02-25 2014-08-11 Asahi Kasei E Materials Corp Copper oxide etching solution and etching method using the same
SG183847A1 (en) 2010-03-04 2012-10-30 Macrogenics Inc Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
US8802091B2 (en) 2010-03-04 2014-08-12 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US10435458B2 (en) 2010-03-04 2019-10-08 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variants with reduced Fcgammar binding
US8642557B2 (en) 2010-03-12 2014-02-04 Abbvie Biotherapeutics Inc. CTLA4 proteins and their uses
TWI667346B (en) 2010-03-30 2019-08-01 中外製藥股份有限公司 Antibodies with modified affinity to fcrn that promote antigen clearance
ES2623799T3 (en) 2010-04-30 2017-07-12 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies and methods for the use of antibodies
BR112012030179A8 (en) 2010-05-27 2023-03-14 Merck Sharp & Dohme FC CONTAINING POLYPEPTIDE
WO2011153243A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Anti-angiogenesis therapy for treating gastric cancer
RU2613886C2 (en) 2010-06-03 2017-03-21 Дженентек, Инк. Antibodies and immunoconjugates rendered by immuno-positron emission tomography, methods of application
MX2020004501A (en) 2010-06-03 2021-11-09 Pharmacyclics Llc The use of inhibitors of bruton's tyrosine kinase (btk).
CA2803588A1 (en) 2010-06-22 2011-12-29 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the c3d fragment of complement component 3
NZ718973A (en) 2010-07-09 2019-01-25 Janssen Vaccines & Prevention Bv Anti-human respiratory syncytial virus (rsv) antibodies and methods of use
NZ605400A (en) 2010-07-09 2015-05-29 Biogen Idec Hemophilia Inc Chimeric clotting factors
US9127057B2 (en) 2010-07-20 2015-09-08 Teva Pharmaceuticals Ausralia Pty Ltd Anti-IL-23 heterodimer specific antibodies
US20130177500A1 (en) 2010-07-23 2013-07-11 Trustee Of Boston University Anti-despr inhibitors as therapeutics for inhibition of pathological angiogenesis and tumor cell invasiveness and for molecular imaging and targeted delivery
BR112013003646B1 (en) 2010-07-29 2021-09-08 Eleven Biotherapeutics, Inc. ISOLATED PROTEIN THAT COMPRISES A DOMAIN OF CYTOKINE OF THE INTERLEUKIN-1 (IL-1) CHIMERIC FAMILY, ITS METHOD OF PREPARATION, PHARMACEUTICAL COMPOSITION AND USES, AS WELL AS, NUCLEIC ACID, VECTOR, AND HOST CELL
ES2667100T3 (en) 2010-08-02 2018-05-09 Macrogenics, Inc. Covalent Diabodies and Their Uses
WO2012019061A2 (en) 2010-08-05 2012-02-09 Stem Centrx, Inc. Novel effectors and methods of use
US20130177555A1 (en) 2010-08-13 2013-07-11 Medimmune Limited Monomeric Polypeptides Comprising Variant FC Regions And Methods Of Use
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
EP2608807A1 (en) 2010-08-27 2013-07-03 Stem Centrx, Inc. Notum protein modulators and methods of use
EP2611464B1 (en) 2010-09-03 2018-04-25 AbbVie Stemcentrx LLC Novel modulators and methods of use
EP2621954A1 (en) 2010-10-01 2013-08-07 Oxford Biotherapeutics Ltd. Anti-rori antibodies
UA112062C2 (en) 2010-10-04 2016-07-25 Бьорінгер Інгельхайм Інтернаціональ Гмбх CD33-Binding Agent
JP6216917B2 (en) 2010-10-13 2017-10-25 ヤンセン バイオテツク,インコーポレーテツド Human oncostatin M antibody and method of use
DK2635607T3 (en) 2010-11-05 2019-11-18 Zymeworks Inc STABLE HETERODIMED ANTIBODY DESIGN WITH MUTATIONS IN THE FC DOMAIN
DK2644698T3 (en) 2010-11-17 2018-01-22 Chugai Pharmaceutical Co Ltd MULTI-SPECIFIC ANTIGEN-BINDING MOLECULE WITH ALTERNATIVE FUNCTION TO BLOOD COAGULATION FACTOR FUNCTION VIII
EP3211009A1 (en) 2010-11-23 2017-08-30 Glaxo Group Limited Antigen binding proteins to oncostatin m (osm)
EP2643351A1 (en) 2010-11-24 2013-10-02 Glaxo Group Limited Multispecific antigen binding proteins targeting hgf
TR201815863T4 (en) 2010-11-30 2018-11-21 Chugai Pharmaceutical Co Ltd Therapeutic agent that induces cytotoxicity.
SG10201509790YA (en) 2010-11-30 2015-12-30 Chugai Pharmaceutical Co Ltd Antigen-Binding Molecule Capable Of Binding To Plurality Of Antigen Molecules Repeatedly
SG190990A1 (en) 2010-12-08 2013-07-31 Stem Centrx Inc Novel modulators and methods of use
EA201390923A1 (en) * 2010-12-22 2013-12-30 Сефалон Острэйлиа Пти Лтд. MODIFIED ANTIBODY WITH AN IMPROVED ELEMENTATION SEMIUM PERIOD
JOP20210044A1 (en) 2010-12-30 2017-06-16 Takeda Pharmaceuticals Co Anti-cd38 antibodies
WO2012103240A2 (en) 2011-01-25 2012-08-02 Eleven Biotherapeutics, Inc. Receptor binding agents
AU2012212066A1 (en) 2011-02-03 2013-08-15 Alexion Pharmaceuticals, Inc. Use of an anti-CD200 antibody for prolonging the survival of allografts
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
RU2620065C2 (en) 2011-02-08 2017-05-22 МЕДИММЬЮН, ЭлЭлСи Antibodies for specific binding of staphylococcus aureus alpha-toxin and methods of application
SA112330278B1 (en) 2011-02-18 2015-10-09 ستيم سينتركس، انك. Novel modulators and methods of use
MX352889B (en) 2011-02-25 2017-12-13 Chugai Pharmaceutical Co Ltd Fcî“riib-specific fc antibody.
WO2012130831A1 (en) 2011-03-29 2012-10-04 Roche Glycart Ag Antibody fc variants
CN103502273A (en) 2011-04-20 2014-01-08 罗氏格黎卡特股份公司 Method and constructs for the pH dependent passage of the blood-brain-barrier
US10654916B2 (en) 2011-04-21 2020-05-19 The Regents Of The University Of California, A California Corporation Compositions and methods for the treatment of neuromyelitis optica
KR102006393B1 (en) 2011-04-29 2019-08-02 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic nuclease compositions and methods
EP2714079B2 (en) 2011-05-21 2019-08-28 MacroGenics, Inc. Deimmunized serum-binding domains and their use for extending serum half-life
WO2012162561A2 (en) 2011-05-24 2012-11-29 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
KR20140028013A (en) 2011-05-25 2014-03-07 머크 샤프 앤드 돔 코포레이션 Method for preparing fc-containing polypeptides having improved properties
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
US9561274B2 (en) 2011-06-07 2017-02-07 University Of Hawaii Treatment and prevention of cancer with HMGB1 antagonists
SI2717898T1 (en) 2011-06-10 2019-07-31 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof
ES2664972T3 (en) 2011-06-10 2018-04-24 Medimmune Limited Pseudomonas anti-Psl binding molecules and uses thereof
DK2718326T3 (en) 2011-06-13 2020-10-26 Csl Ltd ANTIBODIES TO G-CSFR AND USES THEREOF
CA2839917A1 (en) 2011-06-24 2012-12-27 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
RU2727452C1 (en) * 2011-06-28 2020-07-21 Инхибркс, Инк. Fused serpin polypeptides and methods for use thereof
KR20140068861A (en) * 2011-06-28 2014-06-09 인히브릭스 엘엘씨 Wap domain fusion polypeptides and methods of use thereof
US10400029B2 (en) * 2011-06-28 2019-09-03 Inhibrx, Lp Serpin fusion polypeptides and methods of use thereof
ES2640960T3 (en) 2011-06-28 2017-11-07 Oxford Biotherapeutics Ltd. Antibodies for ADP-ribosyl cyclase 2
UA117901C2 (en) 2011-07-06 2018-10-25 Ґенмаб Б.В. Antibody variants and uses thereof
WO2013012733A1 (en) 2011-07-15 2013-01-24 Biogen Idec Ma Inc. Heterodimeric fc regions, binding molecules comprising same, and methods relating thereto
MX349198B (en) 2011-07-15 2017-07-18 Oncomed Pharmaceuticals Inc * Rspo binding agents and uses thereof.
GB201112429D0 (en) 2011-07-19 2011-08-31 Glaxo Group Ltd Antigen-binding proteins with increased FcRn binding
WO2013014092A1 (en) 2011-07-22 2013-01-31 Csl Behring Gmbh Inhibitory anti -factor xii/xiia monoclonal antibodies and their uses
EP2548892A1 (en) 2011-07-22 2013-01-23 CSL Behring GmbH Inhibitory anti-Factor XII/XIIa monoclonal Antibodies and their uses
US20130022551A1 (en) 2011-07-22 2013-01-24 Trustees Of Boston University DEspR ANTAGONISTS AND AGONISTS AS THERAPEUTICS
WO2013014208A2 (en) 2011-07-27 2013-01-31 Glaxo Group Limited Antigen binding constructs
CN104066439A (en) 2011-07-29 2014-09-24 十一生物治疗股份有限公司 Purified proteins
US20130058947A1 (en) 2011-09-02 2013-03-07 Stem Centrx, Inc Novel Modulators and Methods of Use
UY34317A (en) 2011-09-12 2013-02-28 Genzyme Corp T cell antireceptor antibody (alpha) / ß
US20130108641A1 (en) 2011-09-14 2013-05-02 Sanofi Anti-gitr antibodies
EP3485903B1 (en) 2011-09-23 2022-11-16 Mereo BioPharma 5, Inc. Vegf/dll4 binding agents and uses thereof
EA035018B1 (en) 2011-09-30 2020-04-17 Тева Фармасьютикал Австралия Пти Лтд. ANTIBODIES AGAINST TL1a AND USES THEREOF
EP3939996A1 (en) 2011-09-30 2022-01-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
TW201326209A (en) 2011-09-30 2013-07-01 Chugai Pharmaceutical Co Ltd Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
JP6352634B2 (en) 2011-09-30 2018-07-04 中外製薬株式会社 Antigen-binding molecules that induce immune responses to target antigens
TWI580693B (en) 2011-09-30 2017-05-01 中外製藥股份有限公司 Antigen-binding molecule that promotes the disappearance of antigen
EP3617313A1 (en) 2011-10-05 2020-03-04 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting clearance from plasma of antigen comprising saccharide chain receptor-binding domain
KR102434073B1 (en) 2011-10-11 2022-08-18 비엘라 바이오, 인크. Cd40l-specific tn3-derived scaffolds and methods of use thereof
KR102196009B1 (en) 2011-10-25 2021-01-04 프로테나 바이오사이언시즈 리미티드 Antibody formulations and methods
KR102037541B1 (en) 2011-10-28 2019-10-29 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 Polypeptide constructs and uses thereof
US9220774B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of treating cancer by administering anti-GPR49 antibodies
CA2853951A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Antibodies and methods of treating cancer
US10598653B2 (en) 2011-11-01 2020-03-24 Bionomics Inc. Methods of blocking cancer stem cell growth
EP2773667A1 (en) 2011-11-01 2014-09-10 Bionomics, Inc. Anti-gpr49 antibodies
CN109897103A (en) 2011-11-04 2019-06-18 酵活有限公司 There is the antibody design of the stabilization heterodimeric of mutation in Fc structural domain
CN103906533A (en) 2011-11-07 2014-07-02 米迪缪尼有限公司 Multispecific and multivalent binding proteins and uses thereof
JP6182152B2 (en) 2011-11-07 2017-08-16 メディミューン,エルエルシー Combination therapy with anti-Pseudomonas Psl and PcrV binding molecules
WO2013074598A1 (en) * 2011-11-18 2013-05-23 Merck Sharp & Dohme Corp. Fc CONTAINING POLYPEPTIDES HAVING INCREASED ANTI-INFLAMMATORY PROPERTIES AND INCREASED FcRN BINDING
DK2797957T3 (en) 2011-11-23 2019-09-23 Medimmune Llc BINDING MOLECULES SPECIFIC TO HER3 AND APPLICATIONS THEREOF
SG10201609301QA (en) 2011-11-30 2016-12-29 Chugai Pharmaceutical Co Ltd Drug containing carrier into cell for forming immune complex
JP6483442B2 (en) 2011-12-05 2019-03-13 エックス−ボディ インコーポレイテッド PDGF receptor beta-binding polypeptide
RS60499B1 (en) 2011-12-20 2020-08-31 Medimmune Llc Modified polypeptides for bispecific antibody scaffolds
WO2013096221A1 (en) * 2011-12-21 2013-06-27 Amgen Inc. Variant fc-polypeptides with enhanced binding to the neonatal fc receptor
CA2859755C (en) 2011-12-23 2021-04-20 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
EP2793944A4 (en) 2011-12-23 2015-09-02 Nicholas B Lydon Immunoglobulins and variants directed against pathogenic microbes
US9988439B2 (en) 2011-12-23 2018-06-05 Nicholas B. Lydon Immunoglobulins and variants directed against pathogenic microbes
KR102041412B1 (en) * 2011-12-30 2019-11-11 한미사이언스 주식회사 Derivatives of Immunglobulin Fc fragment
AU2013202648B2 (en) 2012-01-10 2016-05-19 Konkuk University Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
SG11201403764XA (en) 2012-01-12 2014-07-30 Biogen Idec Inc Chimeric factor viii polypeptides and uses thereof
EP2623110A1 (en) 2012-01-31 2013-08-07 CSL Behring GmbH Factor XII inhibitors for the treatment of neurological inflammatory disorders
SG11201404751UA (en) * 2012-02-09 2014-09-26 Chugai Pharmaceutical Co Ltd Modified fc region of antibody
PL3564260T3 (en) 2012-02-15 2023-03-06 Bioverativ Therapeutics Inc. Factor viii compositions and methods of making and using same
HUE043537T2 (en) 2012-02-15 2019-08-28 Bioverativ Therapeutics Inc Recombinant factor viii proteins
ES2812849T3 (en) 2012-02-24 2021-03-18 Abbvie Stemcentrx Llc Anti-DLL3 antibodies and procedures for using them
RU2736349C2 (en) 2012-02-24 2020-11-16 Чугаи Сейяку Кабусики Кайся Antigen-binding molecule to accelerate antigen disappearance through fcγriib
AR090352A1 (en) 2012-03-28 2014-11-05 Sanofi Sa ANTIBODIES AGAINST BRADICININE B1 RECEIVER LIGANDS
WO2013155346A1 (en) 2012-04-11 2013-10-17 The Regents Of The University Of California Diagnostic tools for response to 6-thiopurine therapy
US9090694B2 (en) * 2012-04-30 2015-07-28 Janssen Biotech, Inc. ST2L antibody antagonists
US10011660B2 (en) 2012-04-30 2018-07-03 Medimmune, Llc Molecules with reduced effector function and extended half-lives, compositions, and uses thereof
MX2014013637A (en) 2012-05-07 2015-02-05 Sanofi Sa Methods for preventing biofilm formation.
CA2872540A1 (en) 2012-05-10 2013-11-14 Zymeworks Inc. Heteromultimer constructs of immunoglobulin heavy chains with mutations in the fc domain
GB201208370D0 (en) 2012-05-14 2012-06-27 Ucb Pharma Sa Antibodies
WO2013175276A1 (en) 2012-05-23 2013-11-28 Argen-X B.V Il-6 binding molecules
EP3795215A1 (en) 2012-05-30 2021-03-24 Chugai Seiyaku Kabushiki Kaisha Target tissue-specific antigen-binding molecule
US20150353630A1 (en) 2012-05-30 2015-12-10 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for eliminating aggregated antigens
EP2858673A4 (en) 2012-06-06 2016-06-22 Oncomed Pharm Inc Binding agents that modulate the hippo pathway and uses thereof
EP3693000B1 (en) 2012-06-08 2022-03-02 Bioverativ Therapeutics Inc. Procoagulant compounds
CA2875247A1 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Chimeric clotting factors
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
SG10201605703TA (en) 2012-07-06 2016-09-29 Genmab Bv Dimeric protein with triple mutations
WO2014008480A2 (en) 2012-07-06 2014-01-09 Biogen Idec Ma Inc. Cell line expressing single chain factor viii polypeptides and uses thereof
EP3632462A1 (en) 2012-07-06 2020-04-08 Genmab B.V. Dimeric protein with triple mutations
NZ703366A (en) 2012-07-11 2018-03-23 Amunix Operating Inc Factor viii complex with xten and von willebrand factor protein, and uses thereof
MA37794B1 (en) 2012-07-13 2017-07-31 Roche Glycart Ag Anti-vegf / anti-ang-2 bispecific antibodies and their use in the treatment of ocular vascular pathologies
BR112015001690A2 (en) 2012-07-24 2017-11-07 Pharmacyclics Inc mutations associated with resistance to bruton tyrosine kinase inhibitors (btk)
EP2877493B1 (en) 2012-07-25 2018-03-21 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
EP2888279A1 (en) 2012-08-22 2015-07-01 Glaxo Group Limited Anti lrp6 antibodies
JP6501521B2 (en) 2012-08-24 2019-04-17 中外製薬株式会社 FcγRIIb-specific Fc region variant
EP3597747B1 (en) 2012-08-24 2023-03-15 Chugai Seiyaku Kabushiki Kaisha Mouse fcgammarii-specific fc antibody
SI2895513T1 (en) 2012-09-12 2018-11-30 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
US9790268B2 (en) 2012-09-12 2017-10-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
MX2015006973A (en) 2012-12-04 2015-09-28 Oncomed Pharm Inc Immunotherapy with binding agents.
CA2894879A1 (en) 2012-12-19 2014-06-26 Amplimmune, Inc. B7-h4 specific antibodies, and compositions and methods of use thereof
KR20150100715A (en) 2012-12-21 2015-09-02 앰플리뮨, 인크. Anti-h7cr antibodies
KR20200134340A (en) 2013-01-10 2020-12-01 젠맵 비. 브이 Human igg1 fc region variants and uses thereof
CA2898262A1 (en) 2013-01-24 2014-07-31 Glaxosmithkline Intellectual Property Development Limited Tnf-alpha antigen-binding proteins
KR102253597B1 (en) 2013-01-25 2021-05-17 샤이어 휴먼 지네틱 테라피즈 인크. Follistatin in treating duchenne muscular dystrophy
MX2015010023A (en) 2013-02-01 2017-11-17 Transbio Ltd Anti-cd83 antibodies and use thereof.
EP2950885B1 (en) 2013-02-04 2018-11-21 Oncomed Pharmaceuticals, Inc. Methods and monitoring of treatment with a wnt pathway inhibitor
US9340618B2 (en) 2013-02-07 2016-05-17 Csl Limited IL-11R binding proteins
PL2956477T3 (en) 2013-02-15 2021-06-14 Bioverativ Therapeutics Inc. Optimized factor viii gene
BR112015019909A2 (en) 2013-02-22 2017-08-29 Abbvie Stemcentrx Llc ANTIBODY-DRUG CONJUGATES, PHARMACEUTICAL COMPOSITION, THEIR USES, AND KIT
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
AU2014224599B2 (en) 2013-03-08 2018-11-08 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
US9580511B2 (en) 2013-03-11 2017-02-28 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
ES2884813T3 (en) 2013-03-13 2021-12-13 Buzzard Pharmaceuticals AB Chimeric Cytokine Formulations for Ocular Administration
EP2968520B1 (en) 2013-03-14 2021-05-12 MacroGenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor
US20140271641A1 (en) * 2013-03-14 2014-09-18 University Of Guelph Thrombospondin-1 polypeptides and methods of using same
KR101763352B1 (en) 2013-03-15 2017-07-31 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Anti-lag-3 binding proteins
SG10201913874TA (en) 2013-03-15 2020-03-30 Biogen Ma Inc Factor ix polypeptide formulations
CN105451767B (en) 2013-03-15 2019-10-18 泽恩格尼亚股份有限公司 Multivalence and monovalent polyspecific compound and application thereof
MX2015012551A (en) 2013-03-15 2016-10-26 Abbvie Biotechnology Ltd Anti-cd25 antibodies and their uses.
EP2970484B2 (en) 2013-03-15 2022-09-21 Amgen Inc. Heterodimeric bispecific antibodies
US9260527B2 (en) 2013-03-15 2016-02-16 Sdix, Llc Anti-human CXCR4 antibodies and methods of making same
US9587235B2 (en) 2013-03-15 2017-03-07 Atyr Pharma, Inc. Histidyl-tRNA synthetase-Fc conjugates
JP2016514690A (en) 2013-03-15 2016-05-23 アッヴィ バイオテクノロジー リミテッド Anti-CD25 antibodies and their use
ES2699599T3 (en) 2013-03-15 2019-02-11 Abbvie Biotherapeutics Inc Fc variants
US20140302037A1 (en) 2013-03-15 2014-10-09 Amgen Inc. BISPECIFIC-Fc MOLECULES
AU2014250434B2 (en) 2013-04-02 2019-08-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
PT2992013T (en) 2013-04-29 2020-03-05 Teva Pharmaceuticals Australia Pty Ltd Anti-cd38 antibodies and fusions to attenuated interferon alpha-2b
US11117975B2 (en) 2013-04-29 2021-09-14 Teva Pharmaceuticals Australia Pty Ltd Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
EP2999716A2 (en) 2013-05-20 2016-03-30 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
US10513555B2 (en) 2013-07-04 2019-12-24 Prothena Biosciences Limited Antibody formulations and methods
EP3875106A1 (en) 2013-08-08 2021-09-08 Bioverativ Therapeutics Inc. Purification of chimeric fviii molecules
US11384149B2 (en) 2013-08-09 2022-07-12 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
UA116479C2 (en) 2013-08-09 2018-03-26 Макродженікс, Інк. Bi-specific monovalent fc diabodies that are capable of binding cd32b and cd79b and uses thereof
TW201722994A (en) 2013-08-13 2017-07-01 賽諾菲公司 Antibodies to Plasminogen Activator Inhibitor-1 (PAI-1) and uses thereof
UA118267C2 (en) 2013-08-13 2018-12-26 Санофі Antibodies to plasminogen activator inhibitor-1 (pai-1) and uses thereof
TW202003554A (en) 2013-08-14 2020-01-16 美商百歐維拉提夫治療公司 Factor VIII-XTEN fusions and uses thereof
EP2840091A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific diabodies that are capable of binding gpA33 and CD3 and uses thereof
EP2839842A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3 and uses thereof
AU2014312215B2 (en) 2013-08-28 2020-02-27 Abbvie Stemcentrx Llc Site-specific antibody conjugation methods and compositions
CN105792836A (en) 2013-08-28 2016-07-20 施特姆森特克斯股份有限公司 Novel SEZ6 modulators and methods of use
WO2015035044A2 (en) 2013-09-04 2015-03-12 Abbvie Biotherapeutics Inc. Fc VARIANTS WITH IMPROVED ANTIBODY-DEPENDENT CELL-MEDIATED CYTOTOXICITY
PL3702373T3 (en) 2013-09-13 2022-12-05 Beigene Switzerland Gmbh Anti-pd1 antibodies and their use as therapeutics and diagnostics
WO2015048272A1 (en) 2013-09-25 2015-04-02 Amgen Inc. V-c-fc-v-c antibody
ES2900425T3 (en) 2013-09-25 2022-03-16 Bioverativ Therapeutics Inc Column viral inactivation methods
JP6534615B2 (en) 2013-09-27 2019-06-26 中外製薬株式会社 Method for producing polypeptide heteromultimer
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
CN113667012A (en) 2013-10-02 2021-11-19 免疫医疗有限责任公司 Neutralizing anti-influenza a antibodies and uses thereof
DK3055331T3 (en) 2013-10-11 2021-03-22 Oxford Bio Therapeutics Ltd CONJUGATED ANTIBODIES TO LY75 FOR CANCER TREATMENT
WO2015066550A1 (en) 2013-10-31 2015-05-07 Resolve Therapeutics, Llc Therapeutic nuclease-albumin fusions and methods
AU2014346792A1 (en) 2013-11-06 2016-06-02 Abbvie Stemcentrx Llc Novel anti-claudin antibodies and methods of use
EP3065769A4 (en) 2013-11-08 2017-05-31 Biogen MA Inc. Procoagulant fusion compound
GB201320066D0 (en) 2013-11-13 2013-12-25 Ucb Pharma Sa Biological products
EP3074039A4 (en) * 2013-11-26 2017-10-11 The Brigham and Women's Hospital, Inc. Compositions and methods for modulating an immune response
EP3077546A4 (en) 2013-12-02 2017-04-26 Oncomed Pharmaceuticals, Inc. Identification of predictive biomarkers associated with wnt pathway inhibitors
TWI664331B (en) 2013-12-04 2019-07-01 日商中外製藥股份有限公司 Antigen-binding molecules that change antigen-binding ability in response to compound concentration and its database
LT3079719T (en) 2013-12-09 2019-12-10 Allakos Inc Anti-siglec-8 antibodies and methods of use thereof
EP2883883A1 (en) 2013-12-16 2015-06-17 Cardio3 Biosciences S.A. Therapeutic targets and agents useful in treating ischemia reperfusion injury
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
BR112016014810B1 (en) 2013-12-24 2023-12-26 Argenx Bvba FCRN ANTAGONISTS AND METHODS OF USE
CN117106095A (en) 2014-01-10 2023-11-24 比奥贝拉蒂治疗公司 Factor VIII chimeric proteins and uses thereof
US10675352B2 (en) 2014-02-14 2020-06-09 Centrose, Llc Extracellular targeted drug conjugates
EP4014995A1 (en) 2014-02-28 2022-06-22 Allakos Inc. Methods and compositions for treating siglec-8 associated diseases
NZ711451A (en) 2014-03-07 2016-05-27 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
PL3116999T3 (en) 2014-03-14 2021-12-27 F.Hoffmann-La Roche Ag Methods and compositions for secretion of heterologous polypeptides
US9738702B2 (en) * 2014-03-14 2017-08-22 Janssen Biotech, Inc. Antibodies with improved half-life in ferrets
PL3129067T3 (en) 2014-03-19 2023-05-08 Genzyme Corporation Site-specific glycoengineering of targeting moieties
US9885086B2 (en) 2014-03-20 2018-02-06 Pharmacyclics Llc Phospholipase C gamma 2 and resistance associated mutations
WO2015143271A1 (en) 2014-03-21 2015-09-24 X-Body, Inc. Bi-specific antigen-binding polypeptides
CN106536556B (en) 2014-04-04 2020-02-07 生态学有限公司 Humanized antibodies that bind LGR5
LT3128997T (en) 2014-04-08 2020-10-12 Boston Pharmaceuticals Inc. Binding molecules specific for il-21 and uses thereof
UA119352C2 (en) 2014-05-01 2019-06-10 Тева Фармасьютикалз Острейліа Пті Лтд Combination of lenalidomide or pomalidomide and cd38 antibody-attenuated interferon-alpha constructs, and the use thereof
US20170267780A1 (en) 2014-05-16 2017-09-21 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
LT3148579T (en) 2014-05-28 2021-05-25 Agenus Inc. Anti-gitr antibodies and methods of use thereof
JP6675329B2 (en) * 2014-06-12 2020-04-01 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Methods for selecting antibodies with altered FcRn interactions
JP6655074B2 (en) 2014-06-20 2020-02-26 ジェネンテック, インコーポレイテッド Scugacin-based scaffold compositions, methods and uses
EP3160478A4 (en) 2014-06-30 2018-05-16 Bioverativ Therapeutics Inc. Optimized factor ix gene
KR102003754B1 (en) 2014-07-03 2019-07-25 베이진 엘티디 Anti-PD-L1 Antibodies and Their Use as Therapeutics and Diagnostics
JP6837434B2 (en) 2014-07-15 2021-03-03 メディミューン,エルエルシー Neutralization of anti-influenza antibody and its use
CA2955788C (en) 2014-07-22 2024-01-16 Ziyong Sun Anti-pd-1 antibodies
WO2016022630A1 (en) 2014-08-05 2016-02-11 Jiping Zha Anti-pd-l1 antibodies
CA2958018A1 (en) 2014-08-11 2016-02-18 University Of Massachusetts Anti-ospa antibodies and methods of use
WO2016036918A1 (en) 2014-09-03 2016-03-10 Boehringer Ingelheim International Gmbh Compound targeting il-23a and tnf-alpha and uses thereof
TW201617368A (en) 2014-09-05 2016-05-16 史坦森特瑞斯公司 Novel anti-MFI2 antibodies and methods of use
EP3197916A2 (en) 2014-09-25 2017-08-02 Amgen Inc. Protease-activatable bispecific proteins
MA40764A (en) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd THERAPEUTIC AGENT INDUCING CYTOTOXICITY
PE20170702A1 (en) 2014-09-26 2017-06-24 Bayer Pharma AG STABILIZED DERIVATIVES OF ADRENOMEDULIN AND THE USE OF THEM
CN107108721B (en) 2014-09-29 2021-09-07 杜克大学 Bispecific molecules comprising an HIV-1 envelope targeting arm
RU2021125449A (en) 2014-10-01 2021-09-16 Медиммьюн Лимитед ANTIBODIES TO TICAGRELOR AND METHODS OF APPLICATION
CA2964123C (en) 2014-10-09 2023-09-05 Genzyme Corporation Glycoengineered antibody drug conjugates
BR112017007765B1 (en) 2014-10-14 2023-10-03 Halozyme, Inc COMPOSITIONS OF ADENOSINE DEAMINASE-2 (ADA2), VARIANTS THEREOF AND METHODS OF USING THE SAME
US10870704B2 (en) 2014-10-23 2020-12-22 Kira Biotech Pty Limited CD83 binding proteins and uses thereof
EA037749B1 (en) 2014-10-29 2021-05-18 Тева Фармасьютикалз Острэйлиа Пти Лтд INTERFERON 2b VARIANTS
US20160130324A1 (en) 2014-10-31 2016-05-12 Shire Human Genetic Therapies, Inc. C1 Inhibitor Fusion Proteins and Uses Thereof
RU2017119185A (en) 2014-11-05 2018-12-05 Дженентек, Инк. ANTIBODIES AGAINST FGFR2 / 3 AND WAYS OF THEIR APPLICATION
JP6576456B2 (en) * 2014-11-06 2019-09-18 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Fc region variants with modified FcRn binding properties and protein A binding properties
MY191423A (en) 2014-11-10 2022-06-27 Medimmune Ltd Binding molecules specific for cd73 and uses thereof
WO2016075176A1 (en) 2014-11-11 2016-05-19 Medimmune Limited Therapeutic combinations comprising anti-cd73 antibodies and a2a receptor inhibitor and uses thereof
US11008403B2 (en) 2014-11-19 2021-05-18 Genentech, Inc. Anti-transferrin receptor / anti-BACE1 multispecific antibodies and methods of use
US10508151B2 (en) 2014-11-19 2019-12-17 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
HUE052526T2 (en) 2014-11-21 2021-05-28 Bristol Myers Squibb Co Antibodies comprising modified heavy constant regions
TWI758928B (en) 2014-11-21 2022-03-21 美商必治妥美雅史谷比公司 Antibodies against cd73 and uses thereof
US9382321B2 (en) 2014-11-26 2016-07-05 Adventis Health System/Sunbelt, Inc. Effector-deficient anti-CD32A antibodies
WO2016094566A2 (en) 2014-12-10 2016-06-16 Genentech, Inc. Blood brain barrier receptor antibodies and methods of use
TWI779010B (en) 2014-12-19 2022-10-01 日商中外製藥股份有限公司 ANTI-MYOSTATIN ANTIBODIES, POLYPEPTIDES CONTAINING VARIANT Fc REGIONs, AND METHODS OF USE
PT3233921T (en) 2014-12-19 2021-12-09 Chugai Pharmaceutical Co Ltd Anti-c5 antibodies and methods of use
US10940212B2 (en) 2014-12-19 2021-03-09 Monash University IL-21 agonist antibodies and methods of treatment using same
ES2862701T3 (en) 2014-12-22 2021-10-07 Univ Rockefeller Anti-MERTK Agonist Antibodies and Uses Thereof
CA2971732A1 (en) 2014-12-23 2016-06-30 Bristol-Myers Squibb Company Antibodies to tigit
CN107530423B (en) 2015-01-14 2022-04-05 布里格姆及妇女医院股份有限公司 Treatment of cancer with anti-LAP monoclonal antibodies
US20180215801A1 (en) 2015-01-29 2018-08-02 Board Of Trustees Of Michigan State University Cryptic polypeptides and uses thereof
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
WO2016131893A1 (en) 2015-02-18 2016-08-25 Medimmune Limited Incretin fusion polypeptides
AU2016224409B2 (en) 2015-02-27 2021-01-28 Chugai Seiyaku Kabushiki Kaisha Composition for treating IL-6-related diseases
PL3268391T3 (en) 2015-03-09 2022-01-03 Argenx Bvba Methods of reducing serum levels of fc-containing agents using fcrn antagonsits
SG11201707593UA (en) 2015-03-31 2017-10-30 Medimmune Ltd A novel il33 form, mutated forms of il33, antibodies, assays and methods of using the same
FR3034420A1 (en) 2015-03-31 2016-10-07 Lab Francais Du Fractionnement ANTI-CD303 MONOCLONAL ANTIBODIES
JP7082484B2 (en) 2015-04-01 2022-06-08 中外製薬株式会社 Method for Producing Polypeptide Heterogeneous Multimer
LT3303396T (en) 2015-05-29 2023-01-10 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
BR112017025995A2 (en) 2015-06-04 2018-08-14 Ospedale San Raffaele S.R.L. igfbp3 inhibitor, pharmaceutical composition for use in the treatment and / or prevention of an intestinal disorder, method for the diagnosis of an intestinal disorder in an individual and kit for the diagnosis of an intestinal disorder
MX367312B (en) 2015-06-04 2019-08-14 Ospedale San Raffaele Srl Inhibitor of igfbp3/tmem219 axis and diabetes.
US10112990B2 (en) 2015-06-05 2018-10-30 Genentech, Inc. Anti-Tau antibodies and methods of use
TWI773646B (en) 2015-06-08 2022-08-11 美商宏觀基因股份有限公司 Lag-3-binding molecules and methods of use thereof
CA2987797A1 (en) 2015-06-17 2016-12-22 Christopher Robert Bebbington Methods and compositions for treating fibrotic diseases
EA201890162A1 (en) 2015-06-29 2018-07-31 Бристол-Маерс Сквибб Компани ANTIBODIES TO CD40 WITH ENHANCED AGONISTIC ACTIVITY
IL256665B2 (en) 2015-07-23 2023-10-01 Boehringer Ingelheim Int Compound targeting il-23a and b-cell activating factor (baff), nucleic acids encoding same and uses of products comprising them
MD3456346T2 (en) 2015-07-30 2021-11-30 Macrogenics Inc PD-1 and LAG-3 binding molecules and methods of use thereof
WO2017023699A1 (en) 2015-07-31 2017-02-09 Medimmune Limited Methods for treating hepcidin-mediated disorders
BR112018002150A2 (en) 2015-08-03 2018-09-18 Bioverativ Therapeutics Inc factor ix fusion proteins and methods of manufacturing and using them
BR112018003186A2 (en) 2015-09-01 2018-09-25 Agenus Inc. anti-pd-1 antibodies and their methods of use
JP7074341B2 (en) 2015-09-02 2022-05-24 イムテップ エス.アー.エス. Anti-LAG-3 antibody
TWI799366B (en) 2015-09-15 2023-04-21 美商建南德克公司 Cystine knot scaffold platform
US20190022092A1 (en) 2015-09-15 2019-01-24 Acerta Pharma B.V. Therapeutic Combinations of a BTK Inhibitor and a GITR Binding Molecule, a 4-1BB Agonist, or an OX40 Agonist
TWI703158B (en) 2015-09-18 2020-09-01 美商希佛隆公司 Antibodies that specifically bind to tl1a
WO2017053705A1 (en) 2015-09-23 2017-03-30 Oncomed Pharmaceuticals, Inc. Methods and compositions for treatment of cancer
UA126549C2 (en) 2015-10-01 2022-11-02 Хіт Байолоджикс, Інк. Compositions and methods for adjoining type i and type ii extracellular domains as heterologous chimeric proteins
RU2731202C2 (en) 2015-10-08 2020-08-31 Макродженикс, Инк. Combined therapy for cancer treatment
EP3363461A4 (en) 2015-10-12 2019-05-15 Aprogen Kic Inc. Anti-cd43 antibody and use thereof for cancer treatment
US10604577B2 (en) 2015-10-22 2020-03-31 Allakos Inc. Methods and compositions for treating systemic mastocytosis
PL3374398T3 (en) 2015-11-10 2020-08-24 Medimmune, Llc Binding molecules specific for asct2 and uses thereof
WO2017095805A1 (en) 2015-11-30 2017-06-08 Abbvie Inc. ANTI-huLRRC15 ANTIBODY DRUG CONJUGATES AND METHODS FOR THEIR USE
CA3006610A1 (en) 2015-11-30 2017-06-08 Abbvie Inc. Anti-hulrrc15 antibody drug conjugates and methods for their use
KR20180083944A (en) 2015-12-02 2018-07-23 아게누스 인코포레이티드 Antibodies and methods for their use
UA125611C2 (en) 2015-12-14 2022-05-04 Макродженікс, Інк. Bispecific molecules having immunoreactivity in relation to pd-1 and ctla-4, and methods of their use
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
EP3398965A4 (en) 2015-12-28 2019-09-18 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of fc region-containing polypeptide
RU2739208C2 (en) 2015-12-31 2020-12-21 Цзянсу Хэнжуй Медицин Ко., Лтд. Anti-pcsk9 antibody, its antigen-binding domain and medical application thereof
JP2019509714A (en) 2016-01-05 2019-04-11 ジエンス ヘンルイ メデイシンカンパニー リミテッドJiangsu Hengrui Medicine Co.,Ltd. PCSK9 antibody, antigen binding fragment thereof and pharmaceutical use
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes
TW201730212A (en) 2016-02-17 2017-09-01 宏觀基因股份有限公司 ROR1-binding molecules, and methods of use thereof
JP2019509735A (en) 2016-03-04 2019-04-11 シャイアー ヒューマン ジェネティック セラピーズ インコーポレイテッド Recombinant follistatin-Fc fusion protein and use in the treatment of Duchenne muscular dystrophy
US20190284293A1 (en) 2016-03-04 2019-09-19 Bristol-Myers Squibb Company Combination therapy with anti-cd73 antibodies
MX2018010672A (en) 2016-03-04 2019-05-27 Univ Rockefeller Antibodies to cd40 with enhanced agonist activity.
AU2017233658B2 (en) 2016-03-14 2023-09-21 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
US11066464B2 (en) 2016-03-21 2021-07-20 Kymab Limited Anti-malarial antibodies that bind circumsporozoite protein
CA3018081A1 (en) 2016-03-22 2017-09-28 Bionomics Limited Administration of an anti-lgr5 monoclonal antibody
CN109563158B (en) 2016-04-04 2022-08-09 比奥贝拉蒂美国公司 Anti-complement factor BB antibodies and uses thereof
MA45473A (en) 2016-04-04 2019-02-13 Shire Human Genetic Therapies CONJUGATE C1 ESTERASE INHIBITOR AND ITS USES
WO2017180813A1 (en) 2016-04-15 2017-10-19 Macrogenics, Inc. Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
JP2019522960A (en) 2016-04-21 2019-08-22 アッヴィ・ステムセントルクス・エル・エル・シー Novel anti-BMPR1B antibody and method of use
TWI781934B (en) 2016-05-27 2022-11-01 美商艾吉納斯公司 Anti-tim-3 antibodies and methods of use thereof
ES2963807T3 (en) 2016-06-08 2024-04-02 Xencor Inc Treatment of IgG4-related diseases with anti-CD19 antibodies cross-linking to CD32B
DK3478713T3 (en) 2016-06-30 2022-06-20 Prothena Biosciences Ltd Compositions for the treatment of amyloidosis
EP3478830B1 (en) 2016-07-01 2024-04-10 Resolve Therapeutics, LLC Optimized binuclease fusions and methods
WO2018007885A1 (en) 2016-07-05 2018-01-11 Beigene, Ltd. COMBINATION OF A PD-l ANTAGONIST AND A RAF INHIBITOR FOR TREATING CANCER
KR20190039937A (en) 2016-07-08 2019-04-16 스태튼 바이오테크놀로지 비.브이. Anti-ApoC3 antibodies and methods of use thereof
MA45674A (en) 2016-07-15 2019-05-22 Takeda Pharmaceuticals Co METHODS AND MATERIALS FOR EVALUATING A RESPONSE TO PLASMOBLAST AND PLASMOCYTE DEPLÉTION TREATMENTS
JP7241677B2 (en) 2016-07-19 2023-03-17 テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド Anti-CD47 combination therapy
SG11201900616UA (en) 2016-08-02 2019-02-27 Visterra Inc Engineered polypeptides and uses thereof
SG11201900027XA (en) 2016-08-05 2019-02-27 Medimmune Llc Anti-o2 antibodies and uses thereof
SG11201801024XA (en) 2016-08-05 2018-05-30 Chugai Pharmaceutical Co Ltd Therapeutic or preventive compositions for il-8-related diseases
US10538579B2 (en) 2016-08-15 2020-01-21 Board Of Regents, The University Of Texas System Bispecific pertussis antibodies
US20210332112A1 (en) * 2016-08-15 2021-10-28 Board Of Regents, The University Of Texas System Stabilized pertussis antibodies with extended half-life
TWI739887B (en) 2016-08-19 2021-09-21 英屬開曼群島商百濟神州有限公司 Treatment cancers using a combination comprising btk inhibitors
JP2019534858A (en) 2016-09-09 2019-12-05 ジェネンテック, インコーポレイテッド Selective peptide inhibitor of FRIZZLED
US20190270821A1 (en) 2016-09-13 2019-09-05 Humanigen, Inc. Epha3 antibodies for the treatment of pulmonary fibrosis
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
TW202246349A (en) 2016-10-11 2022-12-01 美商艾吉納斯公司 Anti-lag-3 antibodies and methods of use thereof
CN109843917B (en) 2016-10-19 2023-10-03 免疫医疗有限责任公司 anti-O1 antibodies and uses thereof
TW202300515A (en) 2016-10-20 2023-01-01 法商賽諾菲公司 Anti-chikv antibodies and uses thereof
KR20190103147A (en) 2016-10-21 2019-09-04 아디맵 엘엘씨 Anti-Respiratory Cell Fusion Virus Antibodies, and Methods of Producing and Using Them
AU2017345786A1 (en) 2016-10-21 2019-05-23 Adimab, Llc Anti-respiratory syncytial virus antibodies, and methods of their generation and use
JP7265984B2 (en) 2016-10-21 2023-04-27 アディマブ, エルエルシー Anti-respiratory syncytial virus antibodies and methods of their production and use
TWI788307B (en) 2016-10-31 2023-01-01 美商艾歐凡斯生物治療公司 Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
WO2018089628A1 (en) 2016-11-09 2018-05-17 Agenus Inc. Anti-ox40 antibodies, anti-gitr antibodies, and methods of use thereof
AU2017368328A1 (en) 2016-12-02 2019-07-18 Bioverativ Therapeutics Inc. Methods of inducing immune tolerance to clotting factors
CN110520150A (en) 2016-12-02 2019-11-29 比奥维拉迪维治疗股份有限公司 Use the method for chimeric coagulation factor therapies hemophilic arthosis
WO2018104893A1 (en) 2016-12-06 2018-06-14 Glaxosmithkline Intellectual Property Development Limited Alpha4-beta7 antibodies with incrased fcrn binding and/or half-life
BR112019011582A2 (en) 2016-12-07 2019-10-22 Agenus Inc. antibodies and their methods of use
MD3551660T2 (en) 2016-12-07 2024-03-31 Agenus Inc Anti-CTLA-4 antibodies and methods of use thereof
CN117820467A (en) 2016-12-07 2024-04-05 基因泰克公司 anti-TAU antibodies and methods of use
KR20230146126A (en) 2016-12-07 2023-10-18 제넨테크, 인크. Anti-tau antibodies and methods of use
WO2018119246A1 (en) 2016-12-21 2018-06-28 Cephalon, Inc. Antibodies that specifically bind to human il-15 and uses thereof
LT3558391T (en) 2016-12-23 2022-07-11 Immunogen, Inc. Immunoconjugates targeting adam9 and methods of use thereof
MX2019007404A (en) 2016-12-23 2019-09-27 Macrogenics Inc Adam9-binding molecules, and methods of use thereof.
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
TW201837168A (en) 2017-01-06 2018-10-16 美商艾歐凡斯生物治療公司 Expansion of tumor infiltrating lymphocytes (TILS) with tumor necrosis factor receptor superfamily (TNFRSF) agonists and therapeutic combinations of TILS and TNFRSF agonists
US11357841B2 (en) 2017-01-06 2022-06-14 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof
EP3574004A1 (en) * 2017-01-25 2019-12-04 Medimmune, LLC Relaxin fusion polypeptides and uses thereof
EP3573989A4 (en) 2017-01-25 2020-11-18 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
EP3573648B1 (en) 2017-01-25 2023-11-22 Molecular Templates, Inc. Cell-targeting molecules comprising de-immunized, shiga toxin a subunit effectors and cd8+ t-cell epitopes
US11203636B2 (en) 2017-02-01 2021-12-21 Yale University Treatment of existing left ventricular heart failure
JP2020508049A (en) 2017-02-17 2020-03-19 デナリ セラピューティクス インコーポレイテッドDenali Therapeutics Inc. Engineered transferrin receptor binding polypeptide
KR102654105B1 (en) 2017-02-17 2024-04-04 사노피 Multispecific binding molecule with specificity for dystroglycan and laminin-2
US10626169B2 (en) 2017-02-17 2020-04-21 Sanofi Multispecific binding molecules having specificity to dystroglycan and laminin-2
BR112019017628A2 (en) 2017-02-24 2020-07-07 Macrogenics, Inc. cd137 x ta binding molecule, pharmaceutical compositions, use of cd137 x ta binding molecule, cd137 binding molecule, use of cd137 binding molecule, her2 / neu binding molecule, use of her2 binding molecule / neu, and use of a composition
WO2018157163A1 (en) 2017-02-27 2018-08-30 Shattuck Labs, Inc. Vsig8-based chimeric proteins
GB201703876D0 (en) 2017-03-10 2017-04-26 Berlin-Chemie Ag Pharmaceutical combinations
MX2019010802A (en) 2017-03-16 2019-10-30 Medimmune Ltd Anti-par2 antibodies and uses thereof.
UY37651A (en) 2017-03-31 2018-10-31 Swedish Orphan Biovitrum Ab Publ IL-1R-I UNION POLYPEPTIDE
TWI788340B (en) 2017-04-07 2023-01-01 美商必治妥美雅史谷比公司 Anti-icos agonist antibodies and uses thereof
TW201841942A (en) 2017-04-13 2018-12-01 美商艾吉納斯公司 Anti-CD137 antibodies and methods of use thereof
WO2018192974A1 (en) 2017-04-18 2018-10-25 Université Libre de Bruxelles Biomarkers and targets for proliferative diseases
EP3612215A4 (en) 2017-04-20 2021-05-26 aTyr Pharma, Inc. Compositions and methods for treating lung inflammation
AU2018255938A1 (en) 2017-04-21 2019-10-31 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
RS64576B1 (en) 2017-05-01 2023-10-31 Agenus Inc Anti-tigit antibodies and methods of use thereof
WO2018203545A1 (en) 2017-05-02 2018-11-08 国立研究開発法人国立精神・神経医療研究センター Method for predicting and evaluating therapeutic effect in diseases related to il-6 and neutrophils
CN110831628A (en) 2017-05-05 2020-02-21 爱乐科斯公司 Methods and compositions for treating allergic eye diseases
CN110832070A (en) 2017-05-10 2020-02-21 艾欧凡斯生物治疗公司 Expansion of liquid tumor-derived tumor infiltrating lymphocytes and therapeutic uses thereof
CN110914294A (en) 2017-05-12 2020-03-24 夏尔人类遗传性治疗公司 Recombinant follistatin-FC fusion protein and use thereof in treatment of Duchenne muscular dystrophy
JOP20190271A1 (en) 2017-05-24 2019-11-21 Novartis Ag Antibody-cytokine engrafted proteins and methods of use for immune related disorders
KR20200013241A (en) 2017-05-25 2020-02-06 브리스톨-마이어스 스큅 컴퍼니 Antibodies Containing Modified Heavy Chain Constant Regions
GB201709970D0 (en) 2017-06-22 2017-08-09 Kymab Ltd Bispecific antigen-binding molecules
CA3066518A1 (en) 2017-06-26 2019-01-03 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
EP3658184B1 (en) 2017-07-27 2023-09-06 Alexion Pharmaceuticals, Inc. High concentration anti-c5 antibody formulations
CA3072334A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
CA3073537A1 (en) 2017-08-22 2019-02-28 Sanabio, Llc Soluble interferon receptors and uses thereof
EP3672990A1 (en) 2017-08-25 2020-07-01 Five Prime Therapeutics, Inc. B7-h4 antibodies and methods of use thereof
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
CA3080103A1 (en) 2017-10-31 2019-05-09 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
JP2021503885A (en) 2017-11-22 2021-02-15 アイオバンス バイオセラピューティクス,インコーポレイテッド Expanded culture of peripheral blood lymphocytes (PBL) from peripheral blood
ES2939461T3 (en) 2017-11-29 2023-04-24 Csl Ltd Method for treating or preventing ischemia-reperfusion injury
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
EA202091410A1 (en) 2017-12-08 2021-01-13 Ардженкс Бвба APPLICATION OF FCRN ANTAGONISTS FOR TREATMENT OF GENERALIZED MYASTHENIA GRAVIS
US11795226B2 (en) 2017-12-12 2023-10-24 Macrogenics, Inc. Bispecific CD16-binding molecules and their use in the treatment of disease
US20210369775A1 (en) 2017-12-15 2021-12-02 Iovance Biotherapeutics, Inc. Systems and methods for determining the beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof and beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof
EP3498293A1 (en) 2017-12-15 2019-06-19 Institut National De La Sante Et De La Recherche Medicale (Inserm) Treatment of monogenic diseases with an anti-cd45rc antibody
US11802154B2 (en) 2017-12-20 2023-10-31 Alexion Pharmaceuticals, Inc. Humanized anti-CD200 antibodies and uses thereof
WO2019133747A1 (en) 2017-12-27 2019-07-04 Bristol-Myers Squibb Company Anti-cd40 antibodies and uses thereof
FR3076294B1 (en) 2017-12-29 2022-01-28 Lab Francais Du Fractionnement METHOD FOR PURIFYING ANTIBODIES FROM RAW MILK
CA3087699A1 (en) 2018-01-05 2019-07-11 Corvidia Therapeutics, Inc. Methods for treating il-6 mediated inflammation without immunosuppression
EP3737750A4 (en) 2018-01-09 2021-11-17 Synthetic Biologics, Inc. Alkaline phosphatase agents for treatment of neurodevelopmental disorders
CN112739715A (en) 2018-01-12 2021-04-30 武田药品工业株式会社 Subcutaneous administration of anti-CD 38 antibodies
JP2021512126A (en) 2018-02-01 2021-05-13 バイオベラティブ セラピューティクス インコーポレイテッド Use of a lentiviral vector expressing factor VIII
CA3090795A1 (en) 2018-02-13 2019-08-22 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes (tils) with adenosine a2a receptor antagonists and therapeutic combinations of tils and adenosine a2a receptor antagonists
BR112020016400A2 (en) 2018-02-14 2020-12-15 Viela Bio, Inc. ANTIBODIES FOR THYROSINE KINASE 3 RECEPTOR BINDER SIMILAR TO MCDONOUGH FELINE SARCOMA (FMS) (FLT3L) AND THEIR USES FOR THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISEASES
JP7337079B2 (en) 2018-02-15 2023-09-01 マクロジェニクス,インコーポレーテッド Mutant CD3 binding domains and their use in combination therapy for the treatment of disease
SG11202008105RA (en) 2018-03-02 2020-09-29 Five Prime Therapeutics Inc B7-h4 antibodies and methods of use thereof
CN116327926A (en) 2018-03-15 2023-06-27 中外制药株式会社 Anti-dengue virus antibodies with cross-reactivity to Zika virus and methods of use
CA3094174A1 (en) 2018-03-20 2019-09-26 Synthetic Biologics, Inc. Alkaline phosphatase agents for treatment of radiation disorders
PE20201265A1 (en) * 2018-03-21 2020-11-19 Alx Oncology Inc ANTIBODIES AGAINST SIGNAL REGULATORY ALPHA PROTEIN AND METHODS OF USE
WO2019180204A1 (en) 2018-03-23 2019-09-26 Université Libre de Bruxelles Wnt signaling agonist molecules
CN112154156A (en) 2018-03-28 2020-12-29 武田药品工业株式会社 Subcutaneous administration of anti-CD 38 antibodies
CN112154153A (en) 2018-03-28 2020-12-29 百时美施贵宝公司 Interleukin-2/interleukin-2 receptor alpha fusion proteins and methods of use
BR112020023118A2 (en) 2018-05-14 2021-04-13 Werewolf Therapeutics, Inc. ACTIVE ACTIVABLE INTERLEUKIN POLYPEPTIDS 12 AND METHODS OF USE OF THESE
KR20210021467A (en) 2018-05-14 2021-02-26 웨어울프 세라퓨틱스, 인크. Activatable interleukin-2 polypeptide and method of use thereof
CA3100071A1 (en) 2018-05-16 2019-11-21 Csl Limited Soluble complement receptor type 1 variants and uses thereof
US20190375822A1 (en) 2018-05-18 2019-12-12 Bioverativ Therapeutics Inc. Methods of treating hemophilia a
WO2019236417A1 (en) 2018-06-04 2019-12-12 Biogen Ma Inc. Anti-vla-4 antibodies having reduced effector function
TW202016151A (en) 2018-06-09 2020-05-01 德商百靈佳殷格翰國際股份有限公司 Multi-specific binding proteins for cancer treatment
GB201809746D0 (en) 2018-06-14 2018-08-01 Berlin Chemie Ag Pharmaceutical combinations
WO2019241628A1 (en) 2018-06-15 2019-12-19 Proclara Biosciences, Inc. General amyloid interaction motif (gaim)
MX2020013466A (en) 2018-06-26 2021-04-19 Immunogen Inc Immunoconjugates targeting adam9 and methods of use thereof.
EP3817720A2 (en) 2018-07-03 2021-05-12 Bristol-Myers Squibb Company Fgf21 formulations
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
CN112423785A (en) 2018-07-19 2021-02-26 瑞泽恩制药公司 Bispecific anti-BCMAx anti-CD 3 antibodies and uses thereof
CN113227385A (en) 2018-08-09 2021-08-06 比奥维拉迪维治疗股份有限公司 Nucleic acid molecules and their use for non-viral gene therapy
WO2020047328A1 (en) 2018-08-29 2020-03-05 Shattuck Labs, Inc. Combination therapies comprising pd-1-based chimeric proteins
WO2020047462A2 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Methods of treating aging-related disorders
AU2019328313A1 (en) * 2018-08-30 2021-02-25 HCW Biologics, Inc. Single-chain chimeric polypeptides and uses thereof
JP7397874B2 (en) 2018-08-30 2023-12-13 エイチシーダブリュー バイオロジックス インコーポレイテッド Multichain chimeric polypeptides and their uses
WO2020047520A1 (en) * 2018-08-31 2020-03-05 Yale University Enpp1 polypeptides and methods of using same
TW202031273A (en) 2018-08-31 2020-09-01 美商艾歐凡斯生物治療公司 Treatment of nsclc patients refractory for anti-pd-1 antibody
AU2019347751A1 (en) 2018-09-27 2021-04-29 Xilio Development, Inc. Masked cytokine polypeptides
AU2019352017A1 (en) 2018-10-03 2021-05-06 Staten Biotechnology B.V. Antibodies specific for human and cynomolgus ApoC3 and methods of use thereof
TW202029980A (en) 2018-10-26 2020-08-16 美商免疫遺傳股份有限公司 Epcam antibodies, activatable antibodies, and immunoconjugates, and uses thereof
AU2019370339A1 (en) 2018-11-01 2021-06-10 Shandong New Time Pharmaceutical Co., Ltd. Bispecific antibody and use thereof
WO2020096989A1 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients refractory for anti-pd-1 antibody
EP4257195A3 (en) 2018-11-09 2024-01-03 University of Massachusetts Anti-cfae antibodies and methods of use
TW202033555A (en) 2018-11-16 2020-09-16 美商必治妥美雅史谷比公司 Anti-nkg2a antibodies and uses thereof
EP3887397A1 (en) 2018-11-28 2021-10-06 Bristol-Myers Squibb Company Antibodies comprising modified heavy constant regions
EP3892632A4 (en) 2018-12-05 2022-08-17 Bica Therapeutics Inc. Modified product of fc domain of antibody
WO2020118011A1 (en) 2018-12-06 2020-06-11 Alexion Pharmaceuticals, Inc. Anti-alk2 antibodies and uses thereof
TW202039583A (en) 2018-12-07 2020-11-01 瑞士商巴克斯歐塔有限公司 Proteinaceous molecules binding factor ixa and factor x
WO2020115283A1 (en) 2018-12-07 2020-06-11 Baxalta GmbH Bispecific antibodies binding factor ixa and factor x
WO2020114616A1 (en) 2018-12-07 2020-06-11 Tillotts Pharma Ag Topical treatment of immune checkpoint inhibitor induced diarrhoea, colitis or enterocolitis using antibodies and fragments thereof
WO2020128049A1 (en) 2018-12-21 2020-06-25 Kymab Limited Fixaxfx bispecific antibody with common light chain
MX2021007768A (en) 2018-12-26 2021-08-24 Xilio Dev Inc Anti-ctla4 antibodies and methods of use thereof.
KR20210111792A (en) 2018-12-28 2021-09-13 스팍스 테라퓨틱스 인크. Binding molecules specific for claudin 18.2, compositions and methods thereof for treating cancer and other diseases
KR20210113261A (en) 2019-01-04 2021-09-15 리졸브 테라퓨틱스, 엘엘씨 Treatment of Sjogren's Disease Using Nuclease Fusion Proteins
EP3914616A1 (en) 2019-01-23 2021-12-01 Encefa Cd31 competitors and uses thereof
MA55080A (en) 2019-02-26 2022-01-05 Inspirna Inc HIGH AFFINITY ANTI-MERTK ANTIBODIES AND ASSOCIATED USES
JP2022521610A (en) 2019-02-26 2022-04-11 ヤンセン バイオテツク,インコーポレーテツド Combination therapy with bispecific anti-EGFR / C-MET antibody and patient stratification
MX2021010288A (en) 2019-03-01 2021-09-23 Iovance Biotherapeutics Inc Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof.
MX2021010665A (en) 2019-03-05 2021-12-10 Prothena Biosciences Ltd Methods of treating al amyloidosis.
US20220153875A1 (en) 2019-03-19 2022-05-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing antigen-binding domain of which binding activity to antigen is changed depending on mta, and library for obtaining said antigen-binding domain
JP2022521850A (en) 2019-04-03 2022-04-12 ジェンザイム・コーポレーション Anti-alpha beta TCR-binding polypeptide with reduced fragmentation
GB2589049C (en) 2019-04-11 2024-02-21 argenx BV Anti-IgE antibodies
WO2020223565A1 (en) 2019-05-01 2020-11-05 Corvidia Therapeutics, Inc. Anti-il-6 antibody formulation
WO2020230091A1 (en) 2019-05-14 2020-11-19 Janssen Biotech, Inc. Combination therapies with bispecific anti-egfr/c-met antibodies and third generation egfr tyrosine kinase inhibitors
CA3137512A1 (en) 2019-05-14 2020-11-19 Werewolf Therapeutics, Inc. Separation moieties and methods and use thereof
SG11202112010RA (en) 2019-06-07 2021-12-30 Argenx Bvba PHARMACEUTICAL FORMULATIONS OF FcRn INHIBITORS SUITABLE FOR SUBCUTANEOUS ADMINISTRATION
AU2020289587A1 (en) 2019-06-07 2021-12-23 Amgen Inc. Bispecific binding constructs with selectively cleavable linkers
CN113993543A (en) 2019-06-10 2022-01-28 武田药品工业株式会社 Combination therapy with anti-CD 38 antibodies
CA3143584A1 (en) 2019-06-18 2020-12-24 Bayer Aktiengesellschaft Adrenomedullin-analogues for long-term stabilization and their use
CA3143035A1 (en) 2019-06-21 2020-12-24 HCW Biologics, Inc. Multi-chain chimeric polypeptides and uses thereof
US20230018417A1 (en) 2019-07-26 2023-01-19 Shire Human Genetic Therapies, Inc. Recombinant Heme Oxygenase-1 (HO-1) for the Treatment of Sickle Cell Disease
CN114502593A (en) 2019-08-06 2022-05-13 葛兰素史密斯克莱知识产权发展有限公司 Biopharmaceutical compositions and related methods
CN114867751A (en) 2019-08-12 2022-08-05 阿帕特夫研究和发展有限公司 4-1BB and OX40 binding proteins and related compositions and methods, anti-4-1 BB antibodies, anti-OX 40 antibodies
EP4013774A1 (en) 2019-08-13 2022-06-22 Elpis Biopharmaceuticals Engineered interleukin-2 receptor beta agonists
JP2022545741A (en) 2019-08-30 2022-10-28 アジェナス インコーポレイテッド ANTI-CD96 ANTIBODY AND METHODS OF USE THEREOF
US20210113634A1 (en) 2019-09-30 2021-04-22 Bioverativ Therapeutics Inc. Lentiviral vector formulations
CN116478292A (en) 2019-11-15 2023-07-25 恩瑟拉有限责任公司 TMEM219 antibodies and therapeutic uses thereof
US20230039165A1 (en) 2019-11-21 2023-02-09 Enthera S.R.L. Igfbp3 antibodies and therapeutic uses thereof
WO2021105389A1 (en) 2019-11-29 2021-06-03 Kymab Limited Treatment for physiological iron overload
CN114867532A (en) 2019-12-18 2022-08-05 豪夫迈·罗氏有限公司 Bispecific anti-CCL 2 antibodies
TW202140552A (en) 2020-01-08 2021-11-01 比利時商阿根思公司 Methods for treating pemphigus disorders
WO2021158938A1 (en) 2020-02-06 2021-08-12 Bristol-Myers Squibb Company Il-10 and uses thereof
EP3868396A1 (en) 2020-02-20 2021-08-25 Enthera S.R.L. Inhibitors and uses thereof
AU2021225962A1 (en) 2020-02-28 2022-10-20 Genzyme Corporation Modified binding polypeptides for optimized drug conjugation
US11365239B2 (en) 2020-03-20 2022-06-21 Tsb Therapeutics (Beijing) Co., Ltd. Anti-SARS-COV-2 antibodies and uses thereof
EP4126931A1 (en) 2020-03-27 2023-02-08 GlaxoSmithKline Biologicals S.A. Hcvm pentamer binding antibodies
WO2021202463A1 (en) 2020-03-30 2021-10-07 Danisco Us Inc Anti-rsv antibodies
KR20230004686A (en) 2020-04-20 2023-01-06 젠자임 코포레이션 Humanized anti-complement factor Bb antibodies and uses thereof
MX2022013176A (en) 2020-04-24 2022-11-30 Millennium Pharm Inc Anti-cd19 antibodies and uses thereof.
US20230192867A1 (en) 2020-05-15 2023-06-22 Bristol-Myers Squibb Company Antibodies to garp
CR20220646A (en) 2020-05-17 2023-10-23 Astrazeneca Uk Ltd Sars-cov-2 antibodies and methods of selecting and using the same
CA3174236A1 (en) 2020-05-22 2021-11-25 Alwin REITER Ace2-fc fusion proteins and uses thereof
WO2021247812A1 (en) 2020-06-04 2021-12-09 Amgen Inc. Bispecific binding constructs
US20230357341A1 (en) 2020-06-10 2023-11-09 Bica Therapeutics Inc. Fusion protein containing erythropoietin polypeptide
EP4168434A1 (en) 2020-06-17 2023-04-26 MedImmune Limited Heterodimeric relaxin fusions and uses thereof
JP2023532019A (en) * 2020-06-23 2023-07-26 マドン アドバイザーズ エルエルシー Anti-CCR5 Monoclonal Antibody-Based Compositions and Methods
WO2021259227A1 (en) 2020-06-23 2021-12-30 江苏康缘药业股份有限公司 Anti-cd38 antibody and use thereof
CA3165342A1 (en) 2020-06-29 2022-01-06 James Arthur Posada Treatment of sjogren's syndrome with nuclease fusion proteins
WO2022010798A1 (en) 2020-07-06 2022-01-13 Kiromic BioPharma, Inc. Mesothelin isoform binding molecules and chimeric pd1 receptor molecules, cells containing the same and uses thereof
AR122933A1 (en) 2020-07-10 2022-10-19 Novo Nordisk As METHODS TO TREAT CARDIOVASCULAR DISEASE
JP2023536904A (en) 2020-08-06 2023-08-30 バイオベラティブ・ユーエスエイ・インコーポレイテッド Inflammatory Cytokines and Fatigue in Subjects With Complement-Mediated Disease
JP2023537078A (en) 2020-08-10 2023-08-30 アストラゼネカ・ユーケイ・リミテッド SARS-CoV-2 Antibodies for Treating and Preventing COVID-19
MX2023002001A (en) 2020-08-18 2023-03-21 Cephalon Llc Anti-par-2 antibodies and methods of use thereof.
JP2023542049A (en) 2020-09-01 2023-10-04 武田薬品工業株式会社 Interleukin-2 muteins and their uses
BR112023004415A2 (en) 2020-09-11 2023-05-09 Medimmune Ltd B7-H4 BINDING THERAPEUTIC MOLECULES
TW202229312A (en) 2020-09-29 2022-08-01 德商英麥提克生物技術股份有限公司 Amidated peptides and their deamidated counterparts displayed by non-hla-a*02 for use in immunotherapy against different types of cancers
DE102020125457A1 (en) 2020-09-29 2022-03-31 Immatics Biotechnologies Gmbh Amidated peptides and their deamidated counterparts presented by HLA-A*02 molecules for use in immunotherapy against various types of cancer
DE102020125465A1 (en) 2020-09-29 2022-03-31 Immatics Biotechnologies Gmbh Amidated peptides and their deamidated counterparts presented by non-HLA-A*02 molecules for use in immunotherapy against various types of cancer
JP2023546359A (en) 2020-10-06 2023-11-02 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of NSCLC patients with tumor-infiltrating lymphocyte therapy
WO2022076606A1 (en) 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
EP4228616A1 (en) 2020-10-14 2023-08-23 Viridian Therapeutics, Inc. Compositions and methods for treatment of thyroid eye disease
CA3193930A1 (en) 2020-10-29 2022-05-05 Formycon Ag Ace2 fusion proteins and uses thereof
CA3202233A1 (en) 2020-11-18 2022-05-27 Kiromic BioPharma, Inc. Gamma-delta t cell manufacturing processes and chimeric pd1 receptor molecules
AU2021390501A1 (en) 2020-12-01 2023-06-29 Aptevo Research And Development Llc Heterodimeric psma and cd3-binding bispecific antibodies
CN116783217A (en) 2020-12-03 2023-09-19 安进公司 Immunoglobulin constructs with multiple binding domains
CA3201560A1 (en) * 2020-12-08 2022-06-16 David Campbell Half-life extending compositions and methods
CA3201588A1 (en) 2020-12-09 2022-06-16 David Campbell Compositions and methods related to tumor activated antibodies targeting psma and effector cell antigens
JP2024501452A (en) 2020-12-11 2024-01-12 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of cancer patients with tumor-infiltrating lymphocyte therapy in combination with BRAF inhibitors and/or MEK inhibitors
JP2024500403A (en) 2020-12-17 2024-01-09 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of cancer with tumor-infiltrating lymphocytes
WO2022133140A1 (en) 2020-12-17 2022-06-23 Iovance Biotherapeutics, Inc. Treatment with tumor infiltrating lymphocyte therapies in combination with ctla-4 and pd-1 inhibitors
JP2023554456A (en) 2020-12-18 2023-12-27 チューハイ トリノマブ ファーマシューティカル カンパニー リミテッド Molecule that specifically binds to respiratory syncytial virus
EP4271791A2 (en) 2020-12-31 2023-11-08 Iovance Biotherapeutics, Inc. Devices and processes for automated production of tumor infiltrating lymphocytes
TW202241925A (en) 2021-01-15 2022-11-01 德商英麥提克生物技術股份有限公司 Peptides displayed by hla for use in immunotherapy against different types of cancers
AU2022211410A1 (en) 2021-01-22 2023-08-10 Elpis Biopharmaceuticals Anti-pd-l1 monoclonal antibodies and fusion proteins with interleukin-15 (il-15), interleukin-15 receptor 15 alpha or interleukin-2
TW202241508A (en) 2021-01-29 2022-11-01 美商艾歐凡斯生物治療公司 Cytokine associated tumor infiltrating lymphocytes compositions and methods
JP2024508488A (en) 2021-03-01 2024-02-27 エクシリオ デベロップメント, インコーポレイテッド Combination of masked CTLA4 and PD1/PD-L1 antibodies to treat cancer
US20220306743A1 (en) 2021-03-01 2022-09-29 Xilio Development, Inc. Combination of ctla4 and pd1/pdl1 antibodies for treating cancer
WO2022184854A2 (en) 2021-03-03 2022-09-09 Formycon Ag Formulations of ace2 fc fusion proteins
TW202300014A (en) 2021-03-05 2023-01-01 美商艾歐凡斯生物治療公司 Tumor storage and cell culture compositions
EP4308691A1 (en) 2021-03-19 2024-01-24 Iovance Biotherapeutics, Inc. Methods for tumor infiltrating lymphocyte (til) expansion related to cd39/cd69 selection and gene knockout in tils
TW202305118A (en) 2021-03-23 2023-02-01 美商艾歐凡斯生物治療公司 Cish gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
JP2024512029A (en) 2021-03-25 2024-03-18 アイオバンス バイオセラピューティクス,インコーポレイテッド Methods and compositions for T cell co-culture efficacy assays and use with cell therapy products
WO2022204581A2 (en) 2021-03-26 2022-09-29 Scholar Rock, Inc. Tgf-beta inhibitors and use thereof
JP2024513837A (en) 2021-03-31 2024-03-27 バイオベラティブ・ユーエスエイ・インコーポレイテッド Reducing surgery-related hemolysis in patients with cold agglutinin disease
WO2022207785A1 (en) 2021-03-31 2022-10-06 Kymab Limited Antibodies to gfral
WO2022207846A1 (en) 2021-03-31 2022-10-06 Cambridge Enterprise Limited Therapeutic inhibitors of gdf15 signalling
JP2024515066A (en) 2021-04-09 2024-04-04 武田薬品工業株式会社 Antibodies targeting complement factor D and uses thereof
JP2024515189A (en) 2021-04-19 2024-04-05 アイオバンス バイオセラピューティクス,インコーポレイテッド Chimeric costimulatory receptors, chemokine receptors, and their uses in cellular immunotherapy - Patents.com
US20230018888A1 (en) 2021-04-26 2023-01-19 Millennium Pharmaceuticals, Inc. Anti-adgre2 antibodies and uses thereof
US20230111279A1 (en) 2021-04-26 2023-04-13 Millennium Pharmaceuticals, Inc. Anti-clec12a antibodies and uses thereof
WO2022245754A1 (en) 2021-05-17 2022-11-24 Iovance Biotherapeutics, Inc. Pd-1 gene-edited tumor infiltrating lymphocytes and uses of same in immunotherapy
CA3220227A1 (en) 2021-05-28 2022-12-01 Matthew Bruce Combination therapies for treating cancer
WO2022256723A2 (en) 2021-06-03 2022-12-08 Scholar Rock, Inc. Tgf-beta inhibitors and therapeutic use thereof
WO2022263357A1 (en) 2021-06-14 2022-12-22 Argenx Iip Bv Anti-il-9 antibodies and methods of use thereof
WO2022263501A1 (en) 2021-06-18 2022-12-22 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
AU2022299185A1 (en) 2021-06-23 2024-01-25 Scholar Rock, Inc. A myostatin pathway inhibitor in combination with a glp-1 pathway activator for use in treating metabolic disorders
CA3226111A1 (en) 2021-07-22 2023-01-26 Iovance Biotherapeutics, Inc. Method for cryopreservation of solid tumor fragments
WO2023009716A1 (en) 2021-07-28 2023-02-02 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors
KR20240040786A (en) 2021-08-03 2024-03-28 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Biopharmaceutical compositions and stable isotope labeled peptide mapping methods
AU2022331610A1 (en) * 2021-08-16 2024-04-04 Yale University Interleukin-12 variants and methods of use
CA3227716A1 (en) 2021-08-20 2023-02-23 Intervet International B.V. Antibodies and igg fusion proteins with an extended half-life
CA3229448A1 (en) 2021-08-23 2023-03-02 Immunitas Therapeutics, Inc. Anti-cd161 antibodies and uses thereof
TW202323289A (en) 2021-08-31 2023-06-16 日商大正製藥股份有限公司 Anti-growth hormone antibody
WO2023039488A1 (en) 2021-09-09 2023-03-16 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1 talen knockdown
CA3232700A1 (en) 2021-09-24 2023-03-30 Rafael CUBAS Expansion processes and agents for tumor infiltrating lymphocytes
AU2022358522A1 (en) 2021-09-30 2024-03-28 Seagen Inc. B7-h4 antibody-drug conjugates for the treatment of cancer
CA3233953A1 (en) 2021-10-05 2023-04-13 Matthew Bruce Combination therapies for treating cancer
AU2022372894A1 (en) 2021-10-20 2024-04-18 Takeda Pharmaceutical Company Limited Compositions targeting bcma and methods of use thereof
AR127482A1 (en) 2021-10-27 2024-01-31 Iovance Biotherapeutics Inc SYSTEMS AND METHODS TO COORDINATE THE MANUFACTURE OF CELLS FOR PATIENT-SPECIFIC IMMUNOTHERAPY
TW202342095A (en) 2021-11-05 2023-11-01 英商阿斯特捷利康英國股份有限公司 Composition for treatment and prevention of covid-19
WO2023086803A1 (en) 2021-11-10 2023-05-19 Iovance Biotherapeutics, Inc. Methods of expansion treatment utilizing cd8 tumor infiltrating lymphocytes
WO2023091968A1 (en) 2021-11-17 2023-05-25 Disc Medicine, Inc. Methods for treating anemia of kidney disease
WO2023094507A1 (en) 2021-11-24 2023-06-01 Formycon Ag Improved ace2 fusion proteins
WO2023094571A1 (en) 2021-11-25 2023-06-01 Formycon Ag Stabilization of ace2 fusion proteins
WO2023111112A1 (en) 2021-12-15 2023-06-22 Medimmune Limited Treatment using heterodimeric relaxin fusions
WO2023114951A1 (en) 2021-12-17 2023-06-22 Viiv Healthcare Company Combination therapies for hiv infections and uses thereof
WO2023139107A1 (en) 2022-01-18 2023-07-27 argenx BV Galectin-10 antibodies
WO2023147488A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Cytokine associated tumor infiltrating lymphocytes compositions and methods
WO2023147486A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Tumor infiltrating lymphocytes engineered to express payloads
TW202348252A (en) 2022-02-16 2023-12-16 英商梅迪繆思有限公司 Combination therapies for treatment of cancer with therapeutic binding molecules
WO2023168352A1 (en) 2022-03-03 2023-09-07 Yale University Humanized 3e10 antibodies, variants, and antigen binding fragments thereof
WO2023169896A1 (en) 2022-03-09 2023-09-14 Astrazeneca Ab BINDING MOLECULES AGAINST FRα
WO2023170216A1 (en) 2022-03-11 2023-09-14 Astrazeneca Ab A SCORING METHOD FOR AN ANTI-FRα ANTIBODY-DRUG CONJUGATE THERAPY
WO2023196877A1 (en) 2022-04-06 2023-10-12 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2023201369A1 (en) 2022-04-15 2023-10-19 Iovance Biotherapeutics, Inc. Til expansion processes using specific cytokine combinations and/or akti treatment
WO2023209177A1 (en) 2022-04-29 2023-11-02 Astrazeneca Uk Limited Sars-cov-2 antibodies and methods of using the same
WO2023212304A1 (en) 2022-04-29 2023-11-02 23Andme, Inc. Antigen binding proteins
KR20230156844A (en) 2022-05-02 2023-11-14 노보 노르디스크 에이/에스 Novel anti-angptl3 antibodies suitable for high concentration compositions and subcutabeous administration
WO2023220597A1 (en) 2022-05-10 2023-11-16 Elpis Biopharmaceuticals Engineered interleukin-2 receptor beta reduced-binding agonist
WO2023220608A1 (en) 2022-05-10 2023-11-16 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with an il-15r agonist
WO2023242372A1 (en) 2022-06-15 2023-12-21 argenx BV Fcrn/hsa binding molecules and methods of use
US20240117021A1 (en) 2022-06-15 2024-04-11 Bioverativ Usa Inc. Anti-complement c1s antibody formulation
WO2023250507A1 (en) 2022-06-24 2023-12-28 Bioverativ Usa Inc. Methods for treating complement-mediated diseases
WO2024011114A1 (en) 2022-07-06 2024-01-11 Iovance Biotherapeutics, Inc. Devices and processes for automated production of tumor infiltrating lymphocytes
WO2024015830A1 (en) 2022-07-12 2024-01-18 Cytomx Therapeutics, Inc. Epcam immunoconjugates and uses thereof
US20240034780A1 (en) 2022-07-22 2024-02-01 Flagship Pioneering Innovations Vi, Llc Antigen Binding Molecules Targeting Thymic Stromal Lymphopoietin (TSLP)
WO2024026395A1 (en) 2022-07-27 2024-02-01 Cephalon Llc Anti-tl1a antibodies for the treatment of ulcerative colitis and crohn's disease
WO2024026386A1 (en) 2022-07-27 2024-02-01 Cephalon Llc Anti-tl1a antibody formulations
WO2024026494A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Viral particles retargeted to transferrin receptor 1
US20240052051A1 (en) 2022-07-29 2024-02-15 Regeneron Pharmaceuticals, Inc. Anti-tfr:payload fusions and methods of use thereof
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
WO2024030758A1 (en) 2022-08-01 2024-02-08 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies
WO2024038183A1 (en) 2022-08-18 2024-02-22 Immunocore Limited Multi-domain binding molecules
WO2024038193A1 (en) 2022-08-18 2024-02-22 Immunocore Limited Multi-domain binding molecules
WO2024042112A1 (en) 2022-08-25 2024-02-29 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins and uses thereof
WO2024050421A2 (en) 2022-08-30 2024-03-07 Flagship Pioneering Innovations Vi, Llc Antigen binding molecules targeting programmed cell death protein 1 (pd-1)
WO2024062074A1 (en) 2022-09-21 2024-03-28 Sanofi Biotechnology Humanized anti-il-1r3 antibody and methods of use

Family Cites Families (170)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4474893A (en) 1981-07-01 1984-10-02 The University of Texas System Cancer Center Recombinant monoclonal antibodies
US4714681A (en) 1981-07-01 1987-12-22 The Board Of Reagents, The University Of Texas System Cancer Center Quadroma cells and trioma cells and methods for the production of same
DE3378250D1 (en) 1982-04-22 1988-11-24 Ici Plc Continuous release formulations
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
US4741900A (en) 1982-11-16 1988-05-03 Cytogen Corporation Antibody-metal ion complexes
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4666884A (en) 1984-04-10 1987-05-19 New England Deaconess Hospital Method of inhibiting binding of von Willebrand factor to human platelets and inducing interaction of platelets with vessel walls
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5128326A (en) 1984-12-06 1992-07-07 Biomatrix, Inc. Drug delivery systems based on hyaluronans derivatives thereof and their salts and methods of producing same
JP2532858B2 (en) 1985-04-01 1996-09-11 セルテツク リミテツド Transformed myeloma cell line
US4980286A (en) 1985-07-05 1990-12-25 Whitehead Institute For Biomedical Research In vivo introduction and expression of foreign genetic material in epithelial cells
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US4880078A (en) 1987-06-29 1989-11-14 Honda Giken Kogyo Kabushiki Kaisha Exhaust muffler
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
US5336603A (en) 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
JP3095168B2 (en) 1988-02-05 2000-10-03 エル. モリソン,シェリー Antibodies with domain-denaturing constants
US4925648A (en) 1988-07-29 1990-05-15 Immunomedics, Inc. Detection and treatment of infectious and inflammatory lesions
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
DE68927933T2 (en) 1988-09-02 1997-08-14 Dyax Corp PRODUCTION AND SELECTION OF RECOMBINANT PROTEINS WITH DIFFERENT BINDING POINTS
KR900005995A (en) 1988-10-31 1990-05-07 우메모또 요시마사 Modified Interleukin-2 and Method of Making the Same
AU634186B2 (en) 1988-11-11 1993-02-18 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
EP0394827A1 (en) 1989-04-26 1990-10-31 F. Hoffmann-La Roche Ag Chimaeric CD4-immunoglobulin polypeptides
DK0479909T3 (en) 1989-06-29 1997-04-07 Medarex Inc Bispecific reagents for AIDS treatment
US5112946A (en) 1989-07-06 1992-05-12 Repligen Corporation Modified pf4 compositions and methods of use
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
US5436146A (en) 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
AU6430190A (en) 1989-10-10 1991-05-16 Pitman-Moore, Inc. Sustained release composition for macromolecular proteins
WO1991006570A1 (en) 1989-10-25 1991-05-16 The University Of Melbourne HYBRID Fc RECEPTOR MOLECULES
JP2571874B2 (en) 1989-11-06 1997-01-16 アルカーメス コントロールド セラピューティクス,インコーポレイテッド Protein microsphere composition
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
AU7247191A (en) 1990-01-11 1991-08-05 Molecular Affinities Corporation Production of antibodies using gene libraries
US5780225A (en) 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
EP1690935A3 (en) 1990-01-12 2008-07-30 Abgenix, Inc. Generation of xenogeneic antibodies
US5314995A (en) 1990-01-22 1994-05-24 Oncogen Therapeutic interleukin-2-antibody based fusion proteins
EP0521985B1 (en) 1990-03-20 1997-09-24 The Trustees Of Columbia University In The City Of New York Chimeric antibodies with receptor binding ligands in place of their constant region
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5349053A (en) 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
ES2246502T3 (en) 1990-08-29 2006-02-16 Genpharm International, Inc. TRANSGENIC NON-HUMAN ANIMALS ABLE TO PRODUCE HETEROLOGICAL ANTIBODIES.
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
CA2092323A1 (en) 1990-10-01 1992-04-02 George Y. Wu Targeting viruses and cells for selective internalization by cells
ATE175118T1 (en) 1990-10-05 1999-01-15 Medarex Inc TARGETED IMMUNOSTIMULATION WITH BISPECIFIC SUBSTANCES
EP0557300B1 (en) 1990-10-29 1997-11-19 Chiron Corporation Bispecific antibodies, method of production, and uses thereof
EP0564531B1 (en) 1990-12-03 1998-03-25 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
WO1992010591A1 (en) 1990-12-14 1992-06-25 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
DK0503648T3 (en) * 1991-03-12 2000-10-30 Biogen Inc CD2 binding domain of lymphocyte function-associated antigen 3
DE69233367T2 (en) 1991-04-10 2005-05-25 The Scripps Research Institute, La Jolla LIBRARIES OF HETERODIMERIC RECEPTORS BY PHAGEMIDES
IE921342A1 (en) 1991-04-26 1992-11-04 Surface Active Ltd Novel antibodies, and methods for their use
CA2109528A1 (en) 1991-05-01 1992-11-02 Gregory A. Prince A method for treating infectious respiratory diseases
AU668870B2 (en) 1991-05-14 1996-05-23 Targetech, Inc Targeted delivery of genes encoding immunogenic proteins
EP0519596B1 (en) 1991-05-17 2005-02-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
DK0587738T3 (en) 1991-06-05 2000-12-18 Univ Connecticut Destined delivery of genes encoding secretory proteins
AU2238292A (en) 1991-06-14 1993-01-12 Xoma Corporation Microbially-produced antibody fragments and their conjugates
US5844095A (en) 1991-06-27 1998-12-01 Bristol-Myers Squibb Company CTLA4 Ig fusion proteins
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
ATE275198T1 (en) 1991-12-02 2004-09-15 Medical Res Council PRODUCTION OF ANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES SEGMENT LIBRARIES.
US5824307A (en) 1991-12-23 1998-10-20 Medimmune, Inc. Human-murine chimeric antibodies against respiratory syncytial virus
US20020102257A1 (en) 1998-09-21 2002-08-01 Leslie Sid Johnson Human-murine chimeric antibodies against respiratory syncytial virus
WO1993014188A1 (en) 1992-01-17 1993-07-22 The Regents Of The University Of Michigan Targeted virus
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US6004554A (en) 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for targeting the vasculature of solid tumors
US5912015A (en) 1992-03-12 1999-06-15 Alkermes Controlled Therapeutics, Inc. Modulated release from biocompatible polymers
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5447851B1 (en) 1992-04-02 1999-07-06 Univ Texas System Board Of Dna encoding a chimeric polypeptide comprising the extracellular domain of tnf receptor fused to igg vectors and host cells
EP0633943A4 (en) 1992-04-03 1997-05-02 Alexander T Young Gene therapy using targeted viral vectors.
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
CA2118508A1 (en) 1992-04-24 1993-11-11 Elizabeth S. Ward Recombinant production of immunoglobulin-like domains in prokaryotic cells
ES2136664T3 (en) 1992-06-09 1999-12-01 Hoppe Ag CLOSING SYSTEM AND LOCKING ASSEMBLY.
EP0749475A4 (en) 1992-08-26 1997-05-07 Harvard College Use of the cytokine ip-10 as an anti-tumor agent
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
DK0671923T3 (en) 1992-10-09 2001-08-13 Advanced Tissue Sciences Inc Liver reserve cells
EP1024198A3 (en) 1992-12-03 2002-05-29 Genzyme Corporation Pseudo-adenoviral vectors for the gene therapy of haemophiliae
US5441050A (en) 1992-12-18 1995-08-15 Neoprobe Corporation Radiation responsive surgical instrument
EP0679196B1 (en) 1993-01-07 2004-05-26 Sequenom, Inc. Dna sequencing by mass spectrometry
US5934272A (en) 1993-01-29 1999-08-10 Aradigm Corporation Device and method of creating aerosolized mist of respiratory drug
AU691811B2 (en) 1993-06-16 1998-05-28 Celltech Therapeutics Limited Antibodies
WO1995015982A2 (en) 1993-12-08 1995-06-15 Genzyme Corporation Process for generating specific antibodies
PT1231268E (en) 1994-01-31 2005-11-30 Univ Boston BANKS OF POLYCLONE ANTIBODIES
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
GB9415379D0 (en) 1994-07-29 1994-09-21 Smithkline Beecham Plc Novel compounds
CN1117155C (en) 1994-07-29 2003-08-06 史密丝克莱恩比彻姆有限公司 Novel compounds
ATE252894T1 (en) 1995-01-05 2003-11-15 Univ Michigan SURFACE-MODIFIED NANOPARTICLES AND METHODS FOR THEIR PRODUCTION AND USE
DK0805628T3 (en) 1995-01-17 2003-07-14 Brigham & Womens Hospital Receptor specific transepithelial transport of immunogens
US6030613A (en) 1995-01-17 2000-02-29 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
US5747035A (en) 1995-04-14 1998-05-05 Genentech, Inc. Polypeptides with increased half-life for use in treating disorders involving the LFA-1 receptor
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
US6019968A (en) 1995-04-14 2000-02-01 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
EP1709970A1 (en) 1995-04-27 2006-10-11 Abgenix, Inc. Human antibodies against EGFR, derived from immunized xenomice
CA2219486A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5811524A (en) * 1995-06-07 1998-09-22 Idec Pharmaceuticals Corporation Neutralizing high affinity human monoclonal antibodies specific to RSV F-protein and methods for their manufacture and therapeutic use thereof
WO1997007788A2 (en) 1995-08-31 1997-03-06 Alkermes Controlled Therapeutics, Inc. Composition for sustained release of an agent
US5723125A (en) 1995-12-28 1998-03-03 Tanox Biosystems, Inc. Hybrid with interferon-alpha and an immunoglobulin Fc linked through a non-immunogenic peptide
JP2978435B2 (en) 1996-01-24 1999-11-15 チッソ株式会社 Method for producing acryloxypropyl silane
ATE508733T1 (en) 1996-03-04 2011-05-15 Penn State Res Found MATERIALS AND METHODS FOR INCREASE CELLULAR INTERNALIZATION
AU5711196A (en) 1996-03-14 1997-10-01 Human Genome Sciences, Inc. Apoptosis inducing molecule i
DE69731289D1 (en) * 1996-03-18 2004-11-25 Univ Texas IMMUNGLOBULIN-LIKE DOMAIN WITH INCREASED HALF-VALUE TIMES
KR20030096450A (en) 1996-03-22 2003-12-31 휴먼 게놈 사이언시즈, 인코포레이티드 Apoptosis inducing molecule ii
WO1997043316A1 (en) 1996-05-10 1997-11-20 Beth Israel Deaconess Medical Center, Inc. Physiologically active molecules with extended half-lives and methods of using same
US5985309A (en) 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
US5855913A (en) 1997-01-16 1999-01-05 Massachusetts Instite Of Technology Particles incorporating surfactants for pulmonary drug delivery
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
WO1998023289A1 (en) 1996-11-27 1998-06-04 The General Hospital Corporation MODULATION OF IgG BINDING TO FcRn
CA2722378C (en) 1996-12-03 2015-02-03 Amgen Fremont Inc. Human antibodies that bind tnf.alpha.
PT954282E (en) 1997-01-16 2005-06-30 Massachusetts Inst Technology PREPARATION OF PARTICLES FOR INHALATION
US6590079B2 (en) 1997-01-30 2003-07-08 Ixsys, Incorporated Anti-αvβ3 recombinant human antibodies, nucleic acids encoding same
US6277375B1 (en) * 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
BRPI9809391B8 (en) 1997-04-14 2021-05-25 Amgen Res Munich Gmbh process for producing an anti-human antigen receptor, human antibody and pharmaceutical composition
IL132560A0 (en) * 1997-05-02 2001-03-19 Genentech Inc A method for making multispecific antibodies having heteromultimeric and common components
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US6133166A (en) 1997-07-01 2000-10-17 The Procter & Gamble Company Cleaning articles comprising a cellulosic fibrous structure having discrete basis weight regions treated with a high internal phase inverse emulsion
US5994511A (en) * 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
US5989463A (en) 1997-09-24 1999-11-23 Alkermes Controlled Therapeutics, Inc. Methods for fabricating polymer-based controlled release devices
SE512663C2 (en) 1997-10-23 2000-04-17 Biogram Ab Active substance encapsulation process in a biodegradable polymer
CN1174993C (en) 1997-11-03 2004-11-10 人体基因组科学有限公司 VEGI, an inhibitor of angiogenesis and tumor growth
JP2002505086A (en) 1998-02-25 2002-02-19 レキシジェン ファーマシューティカルズ コーポレイション Enhanced circulating half-life of antibody-based fusion proteins
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
PT1068241E (en) 1998-04-02 2007-11-19 Genentech Inc Antibody variants and fragments thereof
AU747231B2 (en) 1998-06-24 2002-05-09 Alkermes, Inc. Large porous particles emitted from an inhaler
AU770555B2 (en) 1998-08-17 2004-02-26 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
US6572856B1 (en) * 1998-09-10 2003-06-03 The University Of Virginia Patent Foundation Methods for the prevention and treatment of cancer using anti-C3b(i) antibodies
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
EP2364997A3 (en) 1999-01-15 2012-07-04 Genentech, Inc. Polypeptide variants with altered effector function
US6531580B1 (en) 1999-06-24 2003-03-11 Ixsys, Inc. Anti-αvβ3 recombinant human antibodies and nucleic acids encoding same
ATE474854T1 (en) * 2000-01-27 2010-08-15 Medimmune Llc RSV NEUTRALIZING ANTIBODIES WITH VERY HIGH AFFINITY
US7229619B1 (en) * 2000-11-28 2007-06-12 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
AU4314801A (en) 2000-02-11 2001-08-20 Lexigen Pharm Corp Enhancing the circulating half-life of antibody-based fusion proteins
EP2341075A1 (en) * 2000-03-01 2011-07-06 MedImmune, LLC Antibodies binding to the f protein of a respiratory syncytial virus (rsv)
IL151348A0 (en) 2000-04-13 2003-04-10 Univ Rockefeller Enhancement of antibody-mediated immune responses
US7179900B2 (en) * 2000-11-28 2007-02-20 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US6855493B2 (en) * 2000-11-28 2005-02-15 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US6818216B2 (en) * 2000-11-28 2004-11-16 Medimmune, Inc. Anti-RSV antibodies
ES2727425T3 (en) * 2000-12-12 2019-10-16 Medimmune Llc Molecules with prolonged half-lives, compositions and uses thereof
US7658921B2 (en) * 2000-12-12 2010-02-09 Medimmune, Llc Molecules with extended half-lives, compositions and uses thereof
JP4229704B2 (en) 2001-04-18 2009-02-25 ダイアックス、コープ Fc region polypeptide binding molecule
US6911321B2 (en) 2001-12-19 2005-06-28 Genentech, Inc. Non-human primate Fc receptors and methods of use
US7219797B2 (en) * 2002-01-14 2007-05-22 Alliance Packaging Llc. Box with insert that extends from a side and that divides the box into compartments and methods for forming and using
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US7425618B2 (en) * 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US7425620B2 (en) 2002-08-14 2008-09-16 Scott Koenig FcγRIIB-specific antibodies and methods of use thereof
CN1705491B (en) 2002-09-27 2013-05-29 赞科股份有限公司 Optimized Fc variants and methods for their generation
US7217797B2 (en) 2002-10-15 2007-05-15 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7365168B2 (en) * 2002-10-15 2008-04-29 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7361740B2 (en) * 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
AU2003286467B2 (en) 2002-10-15 2009-10-01 Abbvie Biotherapeutics Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
CA2545539A1 (en) * 2003-10-15 2005-04-28 Pdl Biopharma, Inc. Alteration of fc-fusion protein serum half-lives by mutagenesis of positions 250, 314 and/or 428 of the heavy chain constant region of ig
EP1812068A4 (en) 2004-10-29 2010-06-09 Medimmune Inc Methods of preventing and treating rsv infections and related conditions
EP1997830A1 (en) 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
US8775090B2 (en) 2008-12-12 2014-07-08 Medimmune, Llc Crystals and structure of a human IgG Fc variant with enhanced FcRn binding
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule

Also Published As

Publication number Publication date
US20030190311A1 (en) 2003-10-09
AU2002248184B2 (en) 2008-01-10
EP3569610A3 (en) 2020-03-18
US7083784B2 (en) 2006-08-01
US20100189718A1 (en) 2010-07-29
JP5444010B2 (en) 2014-03-19
JP2017079771A (en) 2017-05-18
EP1355919A2 (en) 2003-10-29
JP6125949B2 (en) 2017-05-10
ES2649037T3 (en) 2018-01-09
US20140377181A1 (en) 2014-12-25
JP5265446B2 (en) 2013-08-14
JP2014050385A (en) 2014-03-20
US8475792B2 (en) 2013-07-02
US7670600B2 (en) 2010-03-02
US20110311454A1 (en) 2011-12-22
WO2002060919A2 (en) 2002-08-08
US20130272964A1 (en) 2013-10-17
US8323962B2 (en) 2012-12-04
JP4336498B2 (en) 2009-09-30
ATE489395T1 (en) 2010-12-15
EP1355919B1 (en) 2010-11-24
JP2008054679A (en) 2008-03-13
US9562100B2 (en) 2017-02-07
ES2727425T3 (en) 2019-10-16
US20130052135A1 (en) 2013-02-28
JP4336376B2 (en) 2009-09-30
WO2002060919A3 (en) 2003-09-04
US20070122403A1 (en) 2007-05-31
CA2431600A1 (en) 2002-08-08
JP2016020344A (en) 2016-02-04
US8795661B2 (en) 2014-08-05
JP2005501514A (en) 2005-01-20
WO2002060919A9 (en) 2003-01-03
DE60143544D1 (en) 2011-01-05
EP2341060A1 (en) 2011-07-06
EP3569610A2 (en) 2019-11-20
US20060198840A1 (en) 2006-09-07
JP2010154855A (en) 2010-07-15
JP2009261394A (en) 2009-11-12
JP6268127B2 (en) 2018-01-24
PT1355919E (en) 2011-03-02
EP1355919A4 (en) 2005-02-09
EP2354149B1 (en) 2017-08-30
EP2357187A1 (en) 2011-08-17
US7704497B2 (en) 2010-04-27
EP2341060B1 (en) 2019-02-20
US8012476B2 (en) 2011-09-06
EP2354149A1 (en) 2011-08-10
CY1111283T1 (en) 2015-08-05
DK1355919T3 (en) 2011-03-14
AU2002248184C1 (en) 2018-01-04

Similar Documents

Publication Publication Date Title
CA2431600C (en) Molecules with extended half-lives, compositions and uses thereof
AU2002248184A1 (en) Molecules with extended half-lives, compositions and uses thereof
EP2407548A1 (en) Molecules with reduced half-lives, compositions and uses thereof
AU2008201419B2 (en) Molecules with extended half-lives, compositions and uses thereof
AU2016202471B2 (en) Molecules with extended half-lives, compositions and uses thereof
CA2766160A1 (en) Modified molecules with increased affinity for fcrn, compositions, and uses thereof

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20211213