AU2013201195A1 - A method for increasing antibody-dependent cytotoxicity with castanospermine - Google Patents

A method for increasing antibody-dependent cytotoxicity with castanospermine Download PDF

Info

Publication number
AU2013201195A1
AU2013201195A1 AU2013201195A AU2013201195A AU2013201195A1 AU 2013201195 A1 AU2013201195 A1 AU 2013201195A1 AU 2013201195 A AU2013201195 A AU 2013201195A AU 2013201195 A AU2013201195 A AU 2013201195A AU 2013201195 A1 AU2013201195 A1 AU 2013201195A1
Authority
AU
Australia
Prior art keywords
cells
composition
tru
immunoglycoproteins
recombinant
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.)
Abandoned
Application number
AU2013201195A
Inventor
Peter Robert Baum
Erik Stephen ESPLING
Peter Armstrong Thompson
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.)
Aptevo Research and Development LLC
Original Assignee
Emergent Product Development Seattle LLC
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
Priority claimed from AU2007308983A external-priority patent/AU2007308983B2/en
Application filed by Emergent Product Development Seattle LLC filed Critical Emergent Product Development Seattle LLC
Priority to AU2013201195A priority Critical patent/AU2013201195A1/en
Publication of AU2013201195A1 publication Critical patent/AU2013201195A1/en
Assigned to EMERGENT PRODUCT DEVELOPMENT SEATTLE, LLC reassignment EMERGENT PRODUCT DEVELOPMENT SEATTLE, LLC Alteration of Name(s) of Applicant(s) under S113 Assignors: TRUBION PHARMACEUTICALS, INC.
Abandoned legal-status Critical Current

Links

Landscapes

  • Peptides Or Proteins (AREA)

Abstract

A composition comprising recombinant immunoglycoproteins, wherein the recombinant immunoglycoprotein molecules each comprise a binding domain that specifically binds to CD37 and have at least a 5-fold increase in antibody-dependent cellular cytotoxicity (ADCC) as compared to control immunoglycoproteins of the same encoded amino acid sequence produced in Chinese Hamster Ovary (CHO) cells, and wherein at least 60% of the recombinant immunoglycoprotein molecules are non-fucosylated.

Description

Regulation 3.2 Revised 2/98 AUSTRALIA Patents Act, 1990 ORIGINAL COMPLETE SPECIFICATION TO BE COMPLETED BY THE APPLICANT NAME OF APPLICANT: Trubion Pharmaceuticals, Inc. ACTUAL INVENTORS: BAUM, Peter Robert ESPLING, Erik Stephen THOMPSON, Peter Armstrong ADDRESS FOR SERVICE: Peter Maxwell and Associates Level 6 60 Pitt Street SYDNEY NSW 2000 INVENTION TITLE: A METHOD FOR INCREASING ANTIBODY-DEPENDENT CYTOTOXICITY WITH CASTANOSPERMINE DETAILS OF ASSOCIATED APPLICATION NO(S): Divisional of Australian Patent Application No. 2007 308 983 filed on 24 October 2007 The following statement is a full description of this invention including the best method of performing it known to us: m:\docs\20071201 \279049.doc MATERIALS AND METHODS FOR IMPROVED IMMUNOGLYCOPROTEINS Cross-reference to Related Application 100011 This application claims the benefit of prior U.S. provisional application no. 60/853,944 filed October 24, 2006, hereby incorporated by reference in its entirety. Field of the Invention [0002] The invention relates to immunoglycoproteins, including antibodies, that have improved properties, including antibody-dependent cell cytotoxicity and glycosylation patterns, cell culturing methods and media for producing such immunoglycoproteins, and uses of such immunoglycoproteins in treatment of disease. Background [0003] Elimination of targeted cell populations with immunopharmaceuticals is an important therapeutic intervention in several indications. The mechanisms of action used by immunopharmaceuticals to effect such elimination of targeted cells can include complement mediated cellular lysis, activation of apoptotic signaling pathways, blockade of signaling pathways required for survival, and antibody-dependent cellular cytotoxicity (ADCC), also referred to as Fc-dependent cellular cytotoxicity. ADCC is a potent mechanism that is believed to be important for the efficacy of many immunopharmaceuticals. [0004] The mechanism for activation of ADCC involves binding of Fc receptors to immunopharmaceutical molecules that are bound to the surface of the target cell. The binding of Fc receptors to immunopharmaceuticals can be mediated by domains within the constant region of immunoglobulins, such as the CH2 and/or CH3 domains. Different types of constant regions may bind different Fc receptors. Examples include the binding of IgG1 Fc domains to cognate Fc receptors CD16 (FEcyRIlI), CD32 (FcyRII-BI and -B2), and CD64 (FecyRI), IgA Fc domains to the cognate Fc receptor CD89 (FecaRI), and IgE domains to cognate Fc receptors FceRl and CD23. 1oa.
10005] Immunopharmaceutical compositions with enhanced Fc receptor binding may exhibit greater potency in ADCC. Reported methods of achieving this with IgG Fe domains include the introduction of amino acid changes and the modification of carbohydrate structures. Modification of carbohydrate structures may be preferable as amino acid changes in the Fc domain may enhance immunogenicity of a pharmaceutical composition. For immunoglobulin molecules it has been demonstrated that attachment of N-linked carbohydrate to Asn-297 of the CH2 domain is critical for ADCC activity. Its removal enzymatically or through mutation of the N-linked consensus site results in little to no ADCC activity. Some studies have reported that the level of ADCC activity for an immunoglobulin molecule is also dependent on the structure of the carbohydrate, but the actual carbohydrate moieties or structure responsible for ADCC have not yet been elucidated. Still less is known about the optimal carbohydrate structure(s) for ADCC of non-immunoglobulin Fc fusion proteins. [0006J In glycoproteins, carbohydrates may attach to the amide nitrogen atom in the side chain of an asparagine in a tripeptide motif Asn-X-Thr/Ser. This type of glycosylation, termed N-linked glycosylation, commences in the endoplasmic reticulum (ER) with the addition of multiple monosaccharides to a dolichol phosphate to form a 14-residue branched carbohydrate complex. This carbohydrate complex is then transferred to the protein by the oligosaccharyltransferase (OST) complex. Before the glycoprotein leaves the lumen of the ER, three glucose molecules are removed from the 14-residue oligosaccharide. The enzymes ER glucosidase I, ER glucosidase II and ER mannosidase are involved in ER processing. [0007 Subsequently, the polypeptides are transported to the Golgi complex, where the N linked sugar chains are modified in many different ways. In the cis and medial compartments of the Golgi complex, the original 14-saccharide N-linked complex may be trimmed through removal of mannose (Man) residues and elongated through addition of N- acetylglucosamine (GlcNac) and/or fucose (Fuc) residues. The various forms of N-linked carbohydrates generally have in common a pentasaccharide core consisting of three mannose and two N acetylglucosamine residues. Finally, in the trans Golgi, other GlcNac residues can be added, followed by galactose (Gal) and a terminal sialic acid (Sial). Carbohydrate processing in the Golgi complex is called "terminal glycosylation" to distinguish it from "core glycosylation," which takes place in the ER. The final complex carbohydrate units can take on many forms and structures, some of which have two, three or four branches (termed biantennary, 2 triantennary or tetraantennary). A number of enzymes are involved in Golgi processing, including Golgi mannosidases IA, IB and IC, GlcNAc-transferase I, Golgi mannosidase II, GlcNAc-transferase 1I, Galactosyl transferase and Sialyl transferase. [0008] One report has suggested that a crucial carbohydrate determinant of FecyRIIa receptor-mediated ADCC activity is the lack of an alpha- 1,6-fucose moiety added to the core N-linked structure (Shinkawa et al., JBiol Chem. 2003 Jan 31;278(5):3466-73; see also Shields et al., JBiol Chem. 2002 Jul 26;277(30):26733-40). The level of another glycoform, bisected N-linked carbohydrate, has also been proposed to be capable of imparting increased ADCC (Umana et al., Nat BiotechnoL 1999 Feb;17(2):176-80) but there is also contradictory evidence (Shinkawa et al., JBiol Chem. 2003 Jan 31;278(5):3466-73). A potential solution to this contradictory evidence has been suggested by the finding that increased GnTIII in host cells produces immunoglobulin not only with increased bisected sugar but also lacking the core fucose modification (Ferrara et al., Biotechnol Bioeng. 2006 Apr 5;93(5):851-61). This agrees with suggestion that fucose alone has the key role in altering ADCC potency and the association with bisected sugar seen by others reflects a linkage in the two modifications in host cells. However, another report in which in vitro treatment of Rituxan and Herceptin antibodies with GnTIII, to increase bisected sugar, resulted in increased ADCC suggests a direct effect of bisected sugar (Hodoniczky et al., Biotechnol. Prog., 2005 Nov-Dec 21(6):1644-52). However, overexpression of Gnt III at very high levels may be toxic to the cell (Umana et al, Biotechnol Prog. 1998 Mar-Apr;14(2):189-92). [0009] Some proposed methods for producing immunoglobulins with lower fucose content have significant drawbacks for manufacture of a biopharmaceutical drug with an optimal ADCC activity for the therapeutic indication. For example, treatment of immunoglobulins with enzymes that remove fucose residues (fucosidases) involves additional costly manufacturing steps with potentially significant economic and drug consistency risks. Molecular engineering of cell lines to knock-out key enzymes involved in the synthesis of fucosylated glycoproteins require special host strains and in current practice do not allow for "tunable" production of drug with varying ADCC potency to optimize efficacy and safety for a therapeutic use. Generation of a comparison non-enhanced ADCC product is expensive and time consuming. The treatment of cell lines with RNAi or antisense molecules to knock down the level of these key enzymes may have unpredictable off-target effects and would be costly if not impractical to implement at manufacturing scale. 3 [00101 Thus, there continues to exist a need for advantageous methods of preparing immunopharmaceuticals with enhanced ADCC as well as for the improved immunopharmaceuticals produced thereby for therapeutic uses. Summary of the Invention [0011] The invention provides culture media and large scale cell culture methods for improving the properties of immunoglycoproteins, including effector functions such as ADCC, and/or glycosylation patterns such as reduction in fucose content. The invention also provides improved immunoglycoproteins produced by such methods, and uses of such immunoglycoproteins in treatment of disease. [0012] In some embodiments, the invention provides a method for increasing the antibody dependent cytoxicity (ADCC) of immunoglycoprotein molecules produced by a host cell, by growing the host cell in culture medium comprising castanospermine at a concentration between about 25 and about 800 pM, or between about 100 and about 500 AM, or between about 100 and about 400 pM, or between about 100 and about 300 pM. In exemplary embodiments, the ADCC is increased at least 2-fold, 3-fold, 4-fold or 5-fold. [0013] In related embodiments, the invention provides a method for increasing the CD16 binding of immunoglycoprotein molecules produced by a host cell, by growing the host cell in culture medium comprising castanospermine at a concentration between about 25 and about 800 pM, or between about 100 and about 500 pM, or between about 100 and about 400 pM, or between about 100 and about 300 pM. In exemplary embodiments, the CD16 binding is increased by at least 50%, 75%, 100%, 125%, 150%, 175% or 200%. [00141 In the methods of the invention, cell growth, viability and/or density is not significantly affected (e.g. remains at least 80% or higher of untreated cells). The level of immunoglycoprotein production in the culture medium may be at least 100 pgImL, 125 pg/mL, or 150 jig/mL. [0015] In any of the preceding embodiments the culture medium may be essentially serum free, and may include a second carbohydrate modifier. [00161 The invention also contemplates compositions comprising immunoglycoprotein molecules produced by the methods described herein, optionally with a sterile pharmaceutically acceptable carrier or diluent. Such compositions may be administered in 4 methods of killing or inhibiting growth of cancer cells which express on their surface a molecule bound by said immunoglycoprotein molecules, or in methods of depleting cells that express on their surface a molecule bound by said immunoglycoprotein molecules. [0017J Methods of the invention generally involve culturing host cells producing the immunoglycoproteins in culture media containing an appropriate concentration of carbohydrate modifier, e.g. castanospermine, and provide an advantage of improving effector function without significantly affecting cell growth or protein production levels. Exemplary immunoglycoproteins that can be manufactured using the methods of the invention include immunoglobulins and small, modular immunopharmaceutical (SMIPTM) products. Such binding molecules prepared according to the methods of the invention advantageously retain substantially the same properties of binding to target and resulting direct biological activity, but exhibit improved effector-mediated functions. [0018] In one aspect, the invention provides a method for improving the antibody dependent cytoxicity (ADCC) and/or the Fc receptor binding of immunoglycoproteins produced by a host cell. Such methods involve growing the host cell in a volume of at least 750 mL, 1 L, 2L, 3L, 4L, 5L, IOL, 15L, 20L or more of culture medium comprising a carbohydrate modifier, e.g., castanospermine, at a concentration that increases the ADCC activity and/or Fe receptor binding of a composition of immunoglycoprotein molecules produced by the host cell. While the optimal concentration of such carbohydrate modifier, e.g., castanospermine, depends on the potency of the carbohydrate modifier and the relative modulation of ADCC desired, exemplary final concentrations of carbohydrate modifiers in the culture media are less than 800 pM, or less than 750, 700, 650, 600, 550, 500, 450, 400, 350, 300, 250, 200, 150, 125, 100, 90, 80, 70, 60, 50, 40, 30, 20, or 10 pM. [00191 The relative effect on ADCC may be modulated by altering the concentration or duration of the carbohydrate modifier, e.g., castanospermine, applied to the cell culture, providing an additional advantage compared to conventional methods of improving ADCC by altering glycosylation. ADCC activity may be measured and expressed using assays known in the art and in exemplary embodiments increases by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10 fold, 15-fold or 20-fold. 5 [00201 Glycosylation and carbohydrate content is known to affect a variety of immunoglobulin effector-mediated functions, including ADCC, CDC and circulating half life. The data described herein show that the methods of the invention are surprisingly able to provide immunoglycoproteins that exhibit improved ADCC without affecting CDC or half-life. Thus, in exemplary embodiments, ADCC of the immunoglycoprotein molecule composition is increased but other immunoglobulin-type effector functions, such as complement-dependent cytoxicity (CDC) and/or prolonged circulating half-life, remain similar or are not significantly affected (e.g., less than 2-fold increase or decrease, or less than 50%, 40%, 30% , 20% or 10% increase or decrease). [0021] The Fc receptor binding of the composition of immunoglycoprotein molecules may be determined as the relative ratio of carbohydrate modifier-treated immunoglycoprotein molecules, vs. untreated immunoglycoprotein molecules, that bind to CD16. Exemplary assays are described below in the examples. Fc receptor binding in exemplary embodiments increases by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 2-fold, 3-fold, 4 fold, 5-fold or 6-fold. An immunoglycoprotein composition produced by host cells treated with carbohydrate modifier, e.g., castanospermine, according to the invention will bind to CD16 (high and low affinity forms, i.e. V or F at amino acid 158) and/or CD32 a orb and/or CD64 with greater affinity in FcR binding assays than immunoglycoprotein compositions produced by host cells not so treated. This increase in Fc receptor binding affinity is shown herein to correlate to an increase in ADCC activity. [0022] The invention also provides methods for altering the carbohydrate content/glycosylation pattern and/or decreasing the fucose content of immunoglycoproteins by growing the host cell in a volume of at least 750 mL, 1 L, 2L, 3L, 4L, 5L, 1OL, 15L, 20L or more of culture medium comprising a carbohydrate modifier, e.g., castanospermine, at a concentration that decreases the total fucose content and/or alters the glycosylation pattern of a composition of immunoglycoprotein molecules produced by the host cell. Exemplary final concentrations of carbohydrate modifiers, e.g., castanospermine, in the culture media are less than 800 pM, or less than 750, 700, 650, 600, 550, 500, 450, 400, 350, 300, 250, 200, 150, 125, 100, 90, 80, 70, 60, 50, 40, 30, 20, or 10 pM. [0023] The relative effect on fucose content may also be modulated by altering the concentration or duration of the carbohydrate modifier, e.g., castanospermine, applied to the cell culture. The total fucose content of a composition may be expressed as the relative ratio 6 or percentage of non-fucosylated immunoglycoprotein molecules to the total number of immunoglycoprotein molecules in a composition. Exemplary compositions contain at least 30%, 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more non-fucosylated molecules. The fucose content of an immunoglycoprotein composition produced by host cells treated with carbohydrate modifier, e.g., castanospermine, according to the invention will be reduced at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold or 10-fold or more compared to compositions produced by host cells not so treated. [0024] In any of the foregoing methods, the host cells may exhibit high levels of growth during exposure to carbohydrate modifiers, e.g., castanospermine. For example, an exemplary population doubling time of CHO cells producing immunoglycoproteins is about 24 hours; a concentration of carbohydrate modifier according to the invention (e.g. a concentration effective to increase ADCC) is not expected to decrease such doubling time. Ideally, an effective concentration of carbohydrate modifier, e.g., castanospermine, does not reduce cell growth by more than 10, 20, 30, 40, 50, 60 or 70% at a time point 72 hours after addition of the carbohydrate modifier. [00251 In any of the foregoing methods, the host cells may exhibit high levels of protein production during exposure to carbohydrate modifiers, e.g., castanospermine. For example, protein production levels in the presence of an effective concentration of carbohydrate modifier, e.g., castanospermine, may be about 50 pg/mL or higher, or about 75, 100, 125, or 150 pg/mL, or higher. Preferably the host cells exhibit both high levels of growth and high levels of protein production. [0026J Any culture media known in the art, including essentially serum-free culture media, may be used. Fed batch, continuous feed, and other types of culturing methods known in the art may also be used with the methods of the invention. The carbohydrate modifiers may be added to the seed train, to the initial batch culture medium, after a rapid growth phase, or continuously with culture medium (e.g. during continuous feeding). For example, the carbohydrate modifier may be added to an early seed train or feedstock at a lOX or IOX concentration, such that subsequent additions of culture media dilute the concentration of carbohydrate modifier to a level that is still effective in achieving improved ADCC of the recombinant products. Alternatively, the carbohydrate modifier at an effective concentration is included in all culture media added to the cells, obviating the need for dilution. In either case, the carbohydrate modifier is added relatively early in the cell culturing process and an 7 effective concentration is maintained throughout the culturing process in order to optimize homogeneity of the immunoglycoprotein. The effect of carbohydrate modifiers is believed to be long-lasting, and can continue to be observed at least 11-12 days after a one-time addition of carbohydrate modifier. [0027] Exemplary carbohydrate modifiers include core glycosylation inhibitors, terminal glycosylation inhibitors, mannosidase inhibitors, and/or early stage carbohydrate modifiers, and optionally include or exclude fucosylation-specific inhibitors, and are described in more detail below. The invention contemplates that combinations of two or more, or three or more carbohydrate modifiers may provide added benefits. Castanospennine is one specifically contemplated carbohydrate modifier. [0028] In another aspect, the invention provides compositions comprising the immunoglycoprotein molecules produced by any of the foregoing methods, that preferably have a binding affinity Kd of at least 107 M', or at least 108 M", or 109 M' for a target molecule. Such compositions may comprise one or more sterile pharmaceutically acceptable carriers or diluents. [0029] In a further aspect, the invention provides therapeutic methods involving administration of such compositions to subjects that would benefit from such administration, e.g. suffering from a disorder mediated by cells expressing the target molecule, or suffering from a type of cancer in which the cancer cells express the target molecule on their surface. The invention also contemplates use of such compositions in methods of depleting cells expressing the target molecule on their surface. Where the target is CD37, the invention specifically contemplates a method of inhibiting cancer cell growth or destroying cancer cells comprising the step of administering to a subject a composition comprising anti-CD37 SMIP products produced according to the methods of the invention. Similarly, where the target is CD20, the invention specifically contemplates a method of inhibiting cancer cell growth or destroying cancer cells comprising the step of administering to a subject a composition comprising anti-CD20 SMIP products produced according to the methods of the invention. In related embodiments, methods of treating cancer involving arresting or reversing cancer progression are contemplated. The invention further provides methods of treating autoimmune or inflammatory diseases by administering anti-CD37 or anti-CD20 SMIP products produced according to the methods of the invention. In related aspects, the invention contemplates use of the glycoprotein compositions of the invention, optionally 8 comprising a sterile carrier or diluent, in preparation of a medicament for treating any of the diseases or disorders described herein. Immunoglycoproteins [0030 The term "immunoglycoprotein" refers to a glycosylated polypeptide that binds to a target molecule and contains sufficient amino acid sequence derived from a constant region of an immunoglobulin to provide an effector function, preferably ADCC and/or CDC. Exemplary molecules will contain a sequence derived from a CH2 domain of an immunoglobulin, or CH2 and CH3 domains derived from one or more immunoglobulins. Specific subsets of immunoglycoproteins contemplated for production according to the invention include single chain proteins which optionally dimerize through covalent or non covalent associations in the hinge and/or CH3 domains. This subset of single chain proteins excludes the typical tetrameric conformation of immunoglobulins (due to the absence of light chains) but includes Fc-ligand or Fc-soluble receptor fusions. Specific examples of single chain proteins include SMIP products. [00311 SMIP products and methods of producing them have been described previously in co-owned U.S. application no. 10/627,556, and US Patent Publications 2003/133939, 2003/0118592, and 2005/0136049, each of which are incorporated herein by reference in their entirety. Single-Chain Multivalent Binding Proteins with Effector Function are described in International Patent Application No. PCT/US07/71052, filed June 12, 2007 (claiming the benefit of U.S.S.N. 60/813,261, filed June 12, 2006 and 60/853,287, filed October 20, 2006), each of which are incorporated herein by reference in their entirety. SMIP products are novel binding domain-immunoglobulin fusion proteins that feature a binding domain for a cognate structure such as an antigen, a counterreceptor or the like; an IgGI, IgA or IgE hinge region polypeptide or a mutant IgGI hinge region polypeptide having either zero, one or two cysteine residues; and immunoglobulin CH2 and CH3 domains. In one embodiment, the binding domain molecule has one or two cysteine residues. In a related embodiment, it is contemplated that when the binding domain molecule comprises two cysteine residues, the first cysteine, which is typically involved in binding between the heavy chain and light chain variable regions, is not deleted or substituted with an amino acid. SMIPs products are capable of ADCC and/or CDC but may be compromised in their ability to form disulfide-linked multimers. Exemplary SMIP products may have one or more 9 binding regions, such as a binding region of an immunoglobulin superfamily member of a variable light chain and/or variable heavy chain binding region derived from an immunoglobulin. In exemplary embodiments these regions are separated by linker peptides, which may be any linker peptide known in the art to be compatible with domain or region joinder. Exemplary linkers are linkers based on the Gly4Ser linker motif, such as (Gly4Ser)n, where n=3-5. Exemplary SMIP products that can be produced according to the invention include products that bind CD20 or CD37. SMIP products that bind CD20 or CD37 and that comprise specific binding sequences and/or amino acid modifications are described in co-owned, co-pending U.S. application nos. 10/627,556 and 11/493,132, each hereby incorporated by reference in its entirety. [0032] Other examples of immunoglycoproteins include binding domain-Ig fusions, wherein the binding domain may be a non-naturally occurring peptide or a fragment of a naturally occurring ligand or receptor. In the case of receptors, fragments of the extracellular domain are preferred. Exemplary fusions with immunoglobulin or Fc regions include: etanercept which is a fusion protein of sTNFRII with the Fc region (U.S. Pat. No. 5,605,690), alefacept which is a fusion protein of LFA-3 expressed on antigen presenting cells with the Fc region (U.S. Pat. No. 5,914,111), a fusion protein of Cytotoxic T Lymphocyte-associated antigen-4 (CTLA-4) with the Fc region [J. Exp. Med., 181, 1869 (1995)], a fusion protein of interleukin 15 with the Fc region [J. Immunol., 160, 5742 (1998)], a fusion protein of factor VII with the Fc region [Proc. Natl. Acad. Sci. USA, 98, 12180 (2001)], a fusion protein of interleukin 10 with the Fc region [J. Immunol., 154, 5590 (1995)], a fusion protein of interleukin 2 with the Fc region [J. Immunol., 146, 915 (1991)], a fusion protein of CD40 with the Fc region [Surgery, 132, 149 (2002)], a fusion protein of Flt-3 (fis-like tyrosine kinase) with the antibody Fc region [Acta. Haemato., 95, 218 (1996)], a fusion protein of OX40 with the antibody Fc region [J. Leu. Biol., 72, 522 (2002)], other CD molecules [e.g., CD2, CD30 (TNFRSF8), CD95 (Fas), CD106 (VCAM-1), CD137], adhesion molecules [e.g., ALCAM (activated leukocyte cell adhesion molecule), cadherins, ICAM (intercellular adhesion molecule)-], ICAM-2, ICAM-3], cytokine receptors [e.g., interleukin-4R, interleukin-5R, interleukin-6R, interleukin-9R, interleukin- I OR, interleukin-12R, interleukin 13Ral, interleukin- 13Ra2, interleukin-15R, interleukin-21R], chemokines, cell death inducing signal molecules [e.g., B7-Hl, DR6 (Death receptor 6), PD-1 (Programmed death 1), TRAIL RI], costimulating molecules [e.g., B7-1, B7-2, B7-112, ICOS (inducible co stimulator)], growth factors [e.g., ErbB2, ErbB3, ErbB4, HGFR], differentiation-inducing 10 factors (e.g., B7-H3), activating factors (e.g., NKG2D), signal transfer molecules (e.g., gp130). [0033] Yet other examples of immunoglycoproteins include antibodies. The term "antibody" herein is defined to include fully assembled antibodies, monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), antibody fragments that can bind antigen ( e.g., Fab', F'(ab)2, Fv, single chain antibodies, diabodies), and recombinant peptides comprising the forgoing as long as they exhibit the desired antigen binding activity. Multimers or aggregates of intact molecules and/or fragments, including chemically derivatized antibodies, are contemplated. Antibodies of any isotype class or subclass, including IgG, IgM, IgD, IgA, and IgE, IgGI, IgG2, IgG3, IgG4, IgAl and IgA2, are contemplated. Different isotypes have different effector functions; for example, IgG1 and IgG3 isotypes have antibody-dependent cellular cytotoxicity (ADCC) activity. [0034] An "immunoglobulin" or "native antibody" is a tetrameric glycoprotein composed of two identical pairs of polypeptide chains (two "light" and two "heavy" chains). The amino-terminal portion of each chain includes a "variable" ("V") region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. Within this variable region, the "hypervariable" region or "complementarity determining region" (CDR) consists of residues 24-34 (Li), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31 35 (HI), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain as described by Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)] and/or those residues from a hypervariable loop (i.e., residues 26-32 (LI), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (Hi), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain as described by [Chothia et al., J. Mol.Biol. 196: 901-917 (1987)]. [00351 The carboxy-terminal portion of each chain contains a constant region. Light chains have a single domain within the constant region. Thus, light chains have one variable region and one constant region domain. Heavy chains have several domains within the constant region. The heavy chains in IgG, IgA, and IgD antibodies have three constant region domains, which are designated CHI, CH2, and CH3, and the heavy chains in IgM and IgE antibodies have four constant region domains, CHI, CH2, CH3 and CH4. Thus, heavy chains have one variable region and three or four constant regions. 11 10036] The heavy chains of immunoglobulins can also be divided into three functional regions: the Fd region (a fragment comprising VH and CHI, i.e., the two N-terminal domains of the heavy chain), the hinge region, and the Fe region (the "fragment crystallizable" region, derived from constant regions and formed after pepsin digestion). The Fd region in combination with the light chain forms an Fab (the "fragment antigen-binding"). Because an antigen will react stereochemically with the antigen-binding region at the amino terminus of each Fab the IgG molecule is divalent, i.e., it can bind to two antigen molecules. The Fc region contains the domains that interact with immunoglobulin receptors on cells and with the initial elements of the complement cascade. Thus, the Fe fragment is generally considered responsible for the effector functions of an immunoglobulin, such as complement fixation and binding to Fc receptors. [0037] The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations or alternative post-translational modifications that may be present in minor amounts, whether produced from hybridomas or recombinant DNA techniques. Nonlimiting examples of monoclonal antibodies include marine, chimeric, humanized, or human antibodies, or variants or derivatives thereof. [0038 Humanizing or modifying antibody sequence to be more human-like is described in, e.g., Jones et al., Nature 321:522 525 (1986); Morrison et al., Proc. Natl. Acad. Sci., U.S.A., 81:6851 6855 (1984); Morrison and Oi, Adv. Immunol., 44:65 92 (1988); Verhoeyer et al., Science 239:1534 1536 (1988); Padlan, Molec. Immun. 28:489 498 (1991); Padlan, Molec. Immunol. 31(3):169 217 (1994); and Kettleborough, C.A. et al., Protein Eng. 4(7):773 83 (1991); Co, M. S., et al. (1994), J. Inmunol. 152, 2968-2976); Studnicka et al. Protein Engineering 7: 805-814 (1994); each of which is incorporated herein by reference. [00391 One method for isolating human monoclonal antibodies is the use of phage display technology. Phage display is described in e.g., Dower et al., WO 91/17271, McCafferty et al., WO 92/01047, and Caton and Koprowski, Proc. Natl. Acad. Sci. USA, 87:6450-6454 (1990), each of which is incorporated herein by reference. Another method for isolating human monoclonal antibodies uses transgenic animals that have no endogenous immunoglobulin production and are engineered to contain human immunoglobulin loci. See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90:2551 (1993); Jakobovits et al., Nature, 12 362:255-258 (1993); Bruggermann et al., Year in Immuno., 7:33 (1993); WO 91/10741, WO 96/34096, WO 98/24893, or U.S. patent application publication nos. 20030194404, 20030031667 or 20020199213; each incorporated herein by reference. [0040] Antibody fragments may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies. "Antibody fragments" comprise a portion of an intact full length antibody, preferably the antigen binding or variable region of the intact antibody, and include multispecific (bispecific, trispecific, etc.) antibodies formed from antibody fragments. Nonlimiting examples of antibody fragments include Fab, Fab', F(ab')2, Fv [variable region], domain antibody (dAb) [Ward et al., Nature 341:544-546, 1989], complementarity determining region (CDR) fragments, single-chain antibodies (scFv) [Bird et al., Science 242:423-426, 1988, and Huston et al., Proc. Natl. Acad. Sci. USA 85:5879-5883, 1988, optionally including a polypeptide linker; and optionally multispecific, Gruber et al., J. Immunol. 152: 5368 (1994)], single chain antibody fragments, diabodies [EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993)], triabodies, tetrabodies, minibodies [Olafsen, et al., Protein Eng Des Sel. 2004 Apr;] 7(4):315-23], linear antibodies [Zapata et al., Protein Eng.,8(10):1057-1062 (1995)]; chelating recombinant antibodies [Neri et al., J Mol Biol. 246:367-73, 1995], tribodies or bibodies [Schoonjans et al., J Immunol. 165:7050-57, 2000; Willems et al., J Chromatogr B Analyt Technol Biomed Life Sci. 786:161-76, 2003], intrabodies [Biocca, et al, EMBO J. 9:101-108, 1990; Colby et al., Proc Natl Acad Sci U S A. 101:17616-21, 2004], nanobodies [Cortez-Retamozo et al., Cancer Research 64:2853-57, 2004], an antigen-binding-domain immunoglobulin fusion protein, a camelized antibody [Desmyter et al., J. Biol. Chem. 276:26285-90, 2001; Ewert et al., Biochemistry 41:3628-36, 2002; U.S. Patent Publication Nos. 20050136049 and 20050037421], a VHH containing antibody, mimetibodies [U.S. Patent Publication Nos. 20050095700 and 20060127404; WO 04/002424 A2; WO 05/081687 A2], or variants or derivatives thereof, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide, such as a CDR sequence, as long as the antibody retains the desired antigen-binding activity. [0041] The term "variant" when used in connection with antibodies refers to polypeptide sequence of an antibody that contains at least one amino acid substitution, deletion, or insertion in the variable region or the portion equivalent to the variable region, provided that the variant retains the desired target binding affinity or biological activity. In addition, the 13 antibodies of the invention may have amino acid modifications in the constant region to modify effector function of the antibody, including half-life or clearance, ADCC and/or CDC activity. Such modifications can enhance pharmacokinetics or enhance the effectiveness of the antibody in treating cancer, for example. In the case of IgGi, modifications to the constant region, particularly the hinge or CH2 region, may increase or decrease effector function, including ADCC and/or CDC activity. In other embodiments, an IgG2 constant region is modified to decrease antibody-antigen aggregate formation. In the case of IgG4, modifications to the constant region, particularly the hinge region, may reduce the formation of half-antibodies. [0042] The term "derivative" when used in connection with antibodies refers to antibodies covalently modified by conjugation to therapeutic or diagnostic agents, labeling (e.g., with radionuclides or various enzymes), covalent polymer attachment such as pegylation (derivatization with polyethylene glycol) and insertion or substitution by chemical synthesis of non-natural amino acids. Derivatives of the invention will retain the binding properties of underivatized molecules of the invention. Conjugation of cancer-targeting antibodies to cytotoxic agent, for example, radioactive isotopes (e.g., 1131, 1125, Y90 and Re186), chemotherapeutic agents, or toxins, may enhance destruction of cancerous cells. [0043] An immunoglycoprotein that is "specific" for a target molecule binds to that target with a greater affinity than any other target. Immiunoglycoproteins of the invention may have affinities for their targets of a Ka of at least about 104 M-, or alternatively of at least about 101 M-, 106 M-, 107 M-, 10 M-1, 10 M, or 10" M'. Such affinities may be readily determined using conventional techniques, such as by using a BlAcore instrument or by radioimmunoassay using radiolabeled target antigen. Affinity data may be analyzed, for example, by the method of Scatchard et al., Ann N.Y. Acad. Sci., 51:660 (1949). Carbohydrate modifiers [0044] A "carbohydrate modifier" is a small organic compound, preferably of molecular weight < 1000 daltons, that inhibits the activity of an enzyme involved in the addition, removal, or modification of sugars that are part of a carbohydrate attached to a polypeptide. Glycosylation is an extremely complex process that takes place in the endoplasmic reticulum ("core glycosylation") and in the Golgi bodies ("terminal glycosylation"). 14 100451 Other polypeptide-based or polynucleotide-based repressors of glycosylation enzymes, including RNAi or antisense that inhibits activity of early stage carbohydrate modifiers, are useful according to the invention but are excluded from the definition of "carbohydrate modifier." [00461 As used herein, "early stage carbohydrate modifier" refers to an inhibitor of one or more of the glycosylation steps prior to addition of N-acetylglucosamine to mannose, including ER glucosidase I, ER glucosidase II, ER mannosidase, Golgi mannosidase IA, Golgi mannosidase IB, Golgi mannosidase IC and GlcNAc-transferase I. [0047] Subsequent glycosylation steps include Golgi mannosidase II, GlcNAc-transferase II, galactosyl transferase and sialyl transferase, fucosyl transferase, and fucokinase. [0048 Exemplary carbohydrate modifiers include any of the following. Castanospermine is believed to be a glucosidase I and II inhibitor. Deoxyfuconojirimycin is a fucosidase inhibitor. 6-Methyl-tetrahydro-pyran-2H-2,3,4-trioI has been reported in vitro to inhibit phosphorylation of L-fucose, the first step in biosynthesis of GDP-L-Fucose. 6,8a - diepi castanospermine is a reported fucosyltransferase inhibitor. l-N-iminosugars A and B (also known as 1-Butyl-5-methyl-piperidine-3,4-diol hydrochloride and 5-Methyl-piperidine-3,4 diol hydrochloride, respectively) have been reported to be fucosyltransferase inhibitors. Deoxymannojirimycin (DMJ) is an ER mannosidase I inhibitor. Kifunensine (Kf) is an ER mannosidase I inhibitor. Swainsonine (Sw) is an ER mannosidase II inhibitor. Monensin (Mn) is an inhibitor of intracellular protein transport between ER and Golgi that interferes with elongation of core oligosaccharide. [00491 Data described herein show that a variety of glycosidase and/or mannosidase inhibitors provide one or more of desired effects of increasing ADCC activity, increasing Fc receptor binding, and altering glycosylation pattern. 100501 In exemplary embodiments, castanospermine (MW 189.21) is added to the culture medium to a final concentration of about 200 gM (corresponding to about 37.8 pg/mL), or concentration ranges greater than about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, or 150 pM, and up to about 300, 275, 250, 225, 200, 175, 150, 125, 100, 75, 60, or 50 pg/mL. For example, ranges of 10-50, or 50-200, or 50-300, or 100-300, or 150-250 pM are contemplated. 15 [0051] In other exemplary embodiments, DMJ, for example DMJ-HC (MW 199.6) is added to the culture medium to a final concentration of about 200 9M (corresponding to about 32.6 ptg DMJ/mL), or concentration ranges greater than about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, or 150 sM, and up to about 300, 275, 250, 225, 200, 175, 150, 125, 100, 75, 60, or 50 pg/mL. For example, ranges of 10-50, or 50-200, or 50-300, or 100-300, or 150-250 pM are contemplated. 10052] In other exemplary embodiments, kifunensine (MW 232.2) is added to the culture medium to a final concentration of about 10 PM (corresponding to about 2.3 jig/mL), or concentration ranges greater than about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 pM, and up to about 50, 45, 40, 35, 30, 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, or 11 pM. For example, ranges of 1 10, or 1-25, or 1-50, or 5-10, or 5-25, or 5-15 pM are contemplated. Recombinant constructs, cells and culturing methods [0053] As used herein, "host cell" specifically excludes rodent hybridomas but includes any other cell that is capable of glycosylation (i.e. addition of carbohydrate to an amino acid of a polypeptide) and that has been modified through recombinant means to express increased levels of a protein product. Progeny of host cells that retain the recombinant modification and the ability to express the protein product are included within the term "host cell". [00541 Exemplary elements of expression vectors or regulatory sequences may include an origin of replication, a promoter, an operator, or other elements that mediate transcription and translation. Promoters can be constitutive or active and may further be cell type specific, tissue specific, individual cell specific, event specific, temporally specific or inducible. Event specific promoters are active or up regulated only upon the occurrence of an event. In addition to the promoter, repressor sequences, negative regulators, or tissue-specific silencers may be inserted to reduce non-specific expression. Other elements include internal ribosome binding sites, a transcription terminator sequence, including a polyadenylation sequence, splice donor and acceptor sites, and an enhancer, a selectable marker and the like. [0055] The culture medium can include any necessary or desirable ingredients known in the art, such as carbohydrates, including glucose, essential and/or non-essential amino acids, lipids and lipid precursors, nucleic acid precursors, vitamins, inorganic salts, trace elements including rare metals, and/or cell growth factors. The culture medium may be chemically defined or may include serum, plant hydrolysates, or other derived substances. The culture 16 medium may be essentially or entirely serum-free or animal-component free. "Essentially serum-free" means that the medium lacks any serum or contains an insignificant amount of serum. Exemplary supplementary amino acids depleted during cell culture include asparagine, aspartic acid, cysteine, cystine, isoleucine, leucine, tryptophan, and valine. [0056] Commercially available lipids and/or lipid precursors include choline, ethanolamine, or phosphoethanolamine, cholesterol, fatty acids such as oleic acid, linoleic acid, linolenic acid, methyl esters, D-alpha-tocopherol, e.g. in acetate form, stearic acid; myristic acid, palmitic acid, palmitoleic acid; or arachidonic acid. Essential amino acids include Arginine, Histidine, Isoleucine, Leucine, Lysine, Methionine, Phenylalanine, Threonine, Tryptophan and Valine. Non-essential amino acids include Alanine, Asparagine, Aspartate, Cysteine, Glutamate, Glutamine, Glycine, Proline, Serine, and Tyrosine. Commercially available inorganic or trace elements, supplied as appropriate salts, include sodium, calcium, potassium, magnesium, copper, iron, zinc, selenium, molybdenum, vanadium, manganese, nickel, silicon, tin, aluminum, barium, cadmium, chromium, cobalt, germanium, potassium, silver, rubidium, zirconium, fluoride, bromide, iodide and chloride. The medium may also optionally include a nonionic surfactant or surface-active agent to protect the cells from the mixing or aeration. The culture medium may also comprise buffers such as sodium bicarbonate, monobasic and dibasic phosphates, HEPES and/or Tris. The culture medium may also comprise inducers of protein production, such as sodium butyrate, or caffeine. [00571 The invention also provides methods for producing an immunoglycoprotein comprising culturing a host cell in any of the culture media or under any of the conditions described herein. Such methods may further include the step of recovering the immunoglycoprotein from the host cells or culture medium. The carbohydrate modifier may be included in the initial culture medium, or may be added during the initial growth phase or at later phases. When the recombinant protein is secreted into the medium, the medium can be harvested periodically and replaced with fresh medium through several harvest cycles. [0058J Although CHO cells, which are widely used for therapeutic protein production, are preferred, any host cells known in the art to produce glycosylated proteins may be used, including yeast cells, plant cells, plants, insect cells, and mammalian cells. Exemplary yeast cells include Pichia, e.g. P. pastoris, and Saccharomyces e.g. S. cerevisiae, as well as Schizosaccharomyces pombe, Kluyveromyces, K. Zactis, K. fragilis, K. bulgaricus, K. 17 wickeramii, K. waltii, K. drosophilarum, K. thernotolerans, and K. marxianus; K. yarrowia; Trichoderma reesia, Neurospora crassa, Schwanniomyces, Schwanniomyces occidentalis, Neurospora, Penicillium, Totypocladium, Aspergillus, A. nidulans, A. niger, Hansenula, Candida, Kloeckera, Torulopsis, and Rhodotorula. Exemplary insect cells include Autographa californica and Spodoptera frugiperda, and Drosophila. Exemplary mammalian cells include varieties of CHO, BHK, HEK-293, NSO, YB2/3, SP2/0, and human cells such as PER-C6 or HT1080, as well as VERO, HeLa, COS, MDCK, NIH3T3, Jurkat, Saos, PC-12, HCT 116, L929, Ltk-, W138, CVI, TM4, W138, Hep G2, MMT, a leukemic cell line, embryonic stem cell or fertilized egg cell. 100591 The cells may be cultured in any culture system and according to any method known in the art, including T-flasks, spinner and shaker flasks, roller bottles and stirred-tank bioreactors. Anchorage-dependent cells can also be cultivated on microcarrier, e.g. polymeric spheres, that are maintained in suspension in stirred-tank bioreactors. Alternatively, cells can be grown in single-cell suspension. Culture medium may be added in a batch process, e.g. where culture medium is added once to the cells in a single batch, or in a fed batch process in which small batches of culture medium are periodically added. Medium can be harvested at the end of culture or several times during culture. Continuously perfused production processes are also known in the art, and involve continuous feeding of fresh medium into the culture, while the same volume is continuously withdrawn from the reactor. Perfused cultures generally achieve higher cell densities than batch cultures and can be maintained for weeks or months with repeated harvests. Use of immunoglycoproteins [0060] The immunoglycoproteins of the invention are useful as therapeutics to treat diseases mediated by the target molecule, or, for example, as cytolytic agents to kill cancer cells that have the target molecule expressed or associated with the cell surface. [0061] "Treatment" or "treating" refers to either a therapeutic treatment or prophylactic or preventative treatments. A therapeutic treatment may improve at least one symptom of disease in an individual receiving treatment or may delay worsening of a progressive disease in an individual, or prevent onset of additional associated diseases. An improved response is assessed by evaluation of clinical criteria well-known in the art for the disease state. 18 100621 A "therapeutically effective dose" or "effective dose" of an immunoglycoprotein refers to that amount of the compound sufficient to result in amelioration of one or more symptoms of the disease being treated. When applied to an individual active ingredient, administered alone, a therapeutically effective dose refers to that ingredient alone. When applied to a combination, a therapeutically effective dose refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously. The doses may be administered based on weight of the patient, e.g., at a dose of 0.01 to 50 mg/kg, and may be administered on a daily or weekly basis, or every 2 weeks, every 3 weeks, or once a month. [0063] To administer the immunoglycoproteins of the invention to humans or test animals, it is preferable to formulate the molecule in a composition comprising one or more pharmaceutically acceptable carriers or diluents, preferably sterile carriers or diluents if the composition is for parenteral administration. The phrase "pharmaceutically or pharmacologically acceptable" refer to molecular entities and compositions that do not produce allergic, or other adverse reactions when administered using routes well-known in the art, as described below. "Pharmaceutically acceptable carriers" include any and all clinically useful solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. Generally, compositions are also essentially free of pyrogens, as well as other impurities that could be harmful to the recipient. [0064] Immunoglycoproteins may be administered orally, topically, transdermally, parenterally, by inhalation spray, vaginally, rectally, or by intracranial injection. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intracisteral injection, or infusion techniques. Administration by intravenous, intradermal, intramusclar, intramammary, intraperitoneal, intrathecal, retrobulbar, intrapulmonary injection and or surgical implantation at a particular site is contemplated as well. [0065] In one embodiment, administration is performed at the site of a cancer or affected tissue needing treatment by direct injection into the site or via a sustained delivery or sustained release mechanism, which can deliver the formulation internally. For example, biodegradable microspheres or capsules or other biodegradable polymer configurations capable of sustained delivery of a composition (e.g., a soluble polypeptide, antibody, or small molecule) can be included in the formulations of the invention implanted near the cancer. 19 [0066] Therapeutic compositions may also be delivered to the patient at multiple sites. The multiple administrations may be rendered simultaneously or may be administered over a continuous period of time. [0067] Injection of aqueous solutions are preferred. Aqueous compositions can be lyophilized for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins. Any suitable lyophilization and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilization and reconstitution can lead to varying degrees of activity loss and that use levels may have to be adjusted to compensate. [0068] In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. It must be stable under the conditions of manufacture and storage and may be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be desirable to include isotonic agents, for example, sugars or sodium chloride. [0069] In addition, the properties of hydrophilicity and hydrophobicity of the compositions contemplated for use in the invention are well balanced, thereby enhancing their utility for both in vitro and especially in vivo uses, while other compositions lacking such balance are of substantially less utility. Specifically, compositions contemplated for use in the invention have an appropriate degree of solubility in aqueous media which permits absorption and bioavailability in the body, while also having a degree of solubility in lipids which permits the compounds to traverse the cell membrane to a putative site of action. [0070] Also contemplated in the present invention is the administration of an immunoglycoprotein composition in conjunction with a second agent. [0071] As an additional aspect, the invention includes kits or articles of manufacture which comprise one or more compounds or compositions packaged in a manner which facilitates their use to practice methods of the invention. In one embodiment, such a kit includes a immunoglycoprotein described herein, optionally with a second therapeutic agent, packaged 20 in a container such as a sealed bottle or vessel, with a label affixed to the container or included in the package that describes use of the compound or composition in practicing the method. Preferably, the compound or composition is packaged in a unit dosage form. The kit may father include a device suitable for administering the composition according to a specific route of administration or for practicing a screening assay. Preferably, the kit contains a label that describes use of the composition. [0072] The invention further contemplates the use of the immunoglycoproteins of the invention in the manufacture of a medicament for the inhibition or prevention or treatment of a disease, condition, or disorder in a subject characterized or mediated by the target to which the immunoglycoprotein binds. Brief Description of the Drawings [0073] Figure I depicts cell growth of CHO cells expressing TRU-016 grown in cell media with various concentrations of castanosperinine, as shown by cell counts of cells/ml. [0074] Figure 2 depicts cell viability of CHO cells expressing TRU-016 grown in cell media with various concentrations of castanospermine, as shown by % of live cells. [00751 Figure 3 depicts CDI6 binding of TRU-015 produced by cells cultured in the presence of varying concentrations of castanospermine and shows geometric mean fluorescent intensity vs. castanospermine concentration. [0076] Figure 4 depicts CD 16 binding, as shown by geometric mean fluorescent intensity, of TRU-016 produced by cells cultured in the presence of various concentrations of 6,8a diepicastanospermine, swainsonine, or deoxymannojirimycin (DMJ). [00771 Figure 5 depicts CD16 binding, as shown by mean fluorescent intensity, of TRU 016 produced by cells cultured in the presence of varying concentrations of kifunensine. 10078] Figure 6 depicts CD16 binding, as shown by mean fluorescent intensity, of Protein A-purified TRU-016 produced by cells cultured in the presence of varying concentrations of castanospermine. 21 [0079] Figures 7 and 8 depict ADCC of TRU-015 measured using PBMC of high affinity and low affinity donors, respectively, and plots concentration of TRU-0 15 added vs. % specific killing. [0080] Figure 9 depicts ADCC of TRU-016 produced by cells cultured in the presence of varying concentrations of castanospermine, and plots % specific killing vs. concentration of TRU-016 added. [0081] Figure 10 depicts ADCC of TRU-016 produced by cells cultured in the presence of various carbohydrate modifiers, and plots % specific killing vs. concentration of TRU-016 added. [00821 Figure II depicts pharmacokinetic data in mice administered TRU-016 produced by cells cultured in the presence of various carbohydrate modifiers. [00831 Figure 12 depicts CD16 binding of TRU-016 in sera of mice administered the TRU-016 produced by cells treated with various carbohydrate modifiers. [0084] Figure 13 depicts relative tumor volume at 8 days in mice implanted with tumor cells and administered TRU-0 16 produced from cells treated with various carbohydrate modifiers, or untreated cells. [0085] Figure 14 depicts % survival of mice implanted with tumor cells and administered TRU-016 produced from cells treated with various carbohydrate modifiers, or untreated cells. [0086] Figure 15 depicts CDC of TRU-015 produced by cells cultured in the presence of castanospermine, and plots % propidium iodide positive (dead cells) vs. concentration of TRU-015 test protein. [0087] Figure 16 depicts CDC of TRU-016 produced by cells cultured in the presence of various carbohydrate modifiers, and plots % propidium iodide positive (dead cells) vs. concentration of TRU-016 test protein. [0088] Figure 17 depicts relative specific protein production of TRU-01 6 over a range of castanospermine concentrations. [0089] Figure 18 depicts the results of an assay for simultaneous binding of TRU-016 to CD37 and FcyRlIIa (CD 16) over a range of castanospermine concentrations. 22 10090] Figure 19 depicts dose response binding curves ofTRU-016 to CD37-expressing cells for a range of castanospermine concentrations. 100911 Figure 20 depicts ADCC activity curves of TRU-016 over a range of castanospermine concentrations. DETAILED DESCRIPTION OF THE INVENTION EXAMPLES EXAMPLE 1 Production of SMIP Products TRU-016 [0092] CD37-specific SMIPs are described in co-owned U.S. Application No. 10/627,556 and U.S. Patent Publication Nos. 2003/133939, 2003/0118592 and 2005/0136049, each incorporated by reference herein in its entirety. An exemplary SMIP, TRU-01 6, is produced as described below. 10093J TRU-016 [G28-1 scFv VHI1S (SSC-P) H WCH2 WCH3J is a recombinant single chain protein that binds to the CD37 antigen. The nucleotide and amino acid sequences of TRU-01 6 are respectively set out in SEQ ID NOS: I and 2. The binding domain was based on the G28-1 antibody sequence previously disclosed in the patent publications listed in the preceding paragraph, which disclosure is incorporated herein by reference. The binding domain is connected to the effector domain, the CH2 and CH3 domains of human IgGI, through a modified hinge region. TRU-016 exists as a dimer in solution. [0094] TRU-01 6 is produced by recombinant DNA technology in a Chinese hamster ovary (CHO) mammalian cell expression system. TRU-016 SMIPs are purified from CHO culture supernatants by Protein A affinity chromatography. Using dPBS, a 50 mL rProtein A FF sepharose column (GE Healthcare rProtein A Sepharose FF, Catalog # 17-0974-04) is equilibrated at 5.0 mis/min (150 cm/hr) for 1.5 column volumes (CV). The culture supernatant is loaded to the rProtein A Sepharose FF column at a flow rate of 1.7mis/min using the AKTA Explorer 100 Air (GE healthcare AKTA Explorer 100 Air, Catalog # 18 1403-00), capturing the recombinant TRU-0 16. The column is washed with dPBS for 5 Column Volumes (CV), then 1.0 M NaCl, 20mM Sodium Phosphate, pH 6.0, and then with 25 mM NaCl, 25mM NaOAc, pH 5.0. These washing steps remove nonspecifically bound 23 CHO host cell proteins from the rProtein A column that contribute to product precipitation after elution. [0095] The recombinant TRU-016 is eluted from the column with 100mM Glycine, pH 3.5. 1 OmL fractions of the eluted product were recovered and the eluted product was then brought to pH 5.0 with 20% of the eluted volume of 0.5 M 2 -(N-Morpholino)ethanesulfonic acid (MES) pH 6.0. This eluted product is concentrated to approximately 25 mg/mL TRU 016 and filter sterilized. [0096] Purified protein is then subjected to GPC size exclusion chromatography (SEC) to achieve further purification of the TRU-016 (dimer) molecule from higher molecular weight aggregates. Using dPBS, an XK 50/100 column (GE healthcare XK 50/100 empty chromatography column, Catalog # 18-8753-01) containing 1 L of Superdex 200 FF sepharose is equilibrated at 12.6 mls/min (38cm/hr) for 1.5 column volumes (CV). A maximum volume of 54 mIs ( 3% CV) of sample is applied to the column. The column continues to run at 12.6 ml/min and the eluted protein is fractionated in 40 mL fractions. Each fraction is analyzed for product quality using an analytic HPLC, and the eluted fractions are pooled for >95% POI (non-aggregated) TRU-016. This resultant pool is filter sterilized at 0.
2 2pm. The material is then concentrated and formulated with 20 mM sodium phosphate and 240 mM sucrose, at pH 6.0. [0097] An alternative method for purification of the glycovariant is as follows. TRU-016 is purified from CHO culture supernatants by Protein A affinity chromatography. Using dPBS, a 1 mL MabSelect affinity chromatography column (GE Healthcare Hitrap MabSelect, catalog #28-4082-53) is equilibrated at 1.0 m/min for 7 column volumes (CV). The culture supernatant is loaded on to the MabSelect column at a flowrate of 1.0 mL/min using the Akta Explorer 100 Air (GE Healthcare, Akta Explorer 100 Air, catalog # 18-1403-00) capturing the recombinant TRU-016. The column is washed with dPBS for 20 CV, then with 20mM Sodium Phosphate, 1.0 M NaCl, pH 7.0 for 5 CV and then with dPBS for 3 CV. 100981 The recombinant TRU-016 is eluted from the column with 10 mM Citrate, pH 3.5 and the column is stripped with 10 mM Citrate 3.0 for 8 CV. Following the strip the column is re-equilibrated for 5 CV with dPBS. The protein is collected into fractions during elution which are pooled based upon absorbance and this pooled material is brought to pH 5.0 with an addition of approximately 400 pL of 0.55 M 2 -(N-Morpholin)ethanesulfonic acid (MES) 24 pH 6.0 per 5 mL of elution. This neutralized eluate is filter sterilized and submitted for both activity assays as well as process analytical assays. [0099J Experiments may be performed to confirm that the binding specificity of the parent antibody to the CD37 cell surface receptor is preserved in TRU-016. Human PBMCs are isolated over LSM density gradients and incubated with unconjugated TRU-016 and PE conjugated anti-human CD 19. Cells are washed and incubated with 1:100 FITC GAH IgG (Fc specific) for 45 minutes on ice. Cells are washed and analyzed by two-color flow cytometry on a FACsCalibur instrument using Cell Quest software. Cells are gated for B lymphocytes or non-B lymphocytes by CD19 staining. [01001 With increasing concentrations of TRU-016, the FITC signal on the B lymphocyte (CD 19 positive gate) increases rapidly from 0.01-1.0 pg/ml, until reaching saturation at approximately 1 pg/mL or a mean fluorescence intensity (MFI) of 1000. In contrast, the staining of the non-B lymphocyte population is detectable, but very low, and increases slowly with increasing concentration of scFvIg. TRU-015 [01011 CD20-specific SMIPs are prepared similarly. CD20-specific SMIPs are described in co-owned US Patent Publications 2003/133939, 2003/0118592 and 2005/0136049, each incorporated by reference herein in its entirety. An exemplary SMIP, TRU-015, is described below. [0102] TRU-015 is a recombinant single chain protein that binds to the CD20 antigen. The nucleotide and amino acid sequences of TRU-015 are respectively set out in SEQ ID NOS: 3 and 4. The binding domain was based on a publicly available human CD20 antibody sequence. The binding domain is connected to the effector domain, the CH2 and CH3 domains of human IgG1, through a modified CSS hinge region. TRU-015 exists as a dimer in solution. (0103] TRU-015 comprises the 2e12 leader peptide cloning sequence from amino acids I 23 of SEQ ID NO: 4; the 2H7 murine anti-human CD20 light chain variable region with a lysine to serine (VHLl IS) amino acid substitution at residue 11 in the variable region, which is reflected at position 34 in SEQ ID NO: 4; an asp-gly 3 -ser-(gly 4 ser) 2 linker, beginning at residue 129 in SEQ ID NO: 4; the 2H7 murine anti-human CD20 heavy chain variable 25 region, which lacks a serine residue at the end of the heavy chain region, i.e., changed from VTVSS to VTVS; a human IgG FEc domain, including a modified hinge region comprising a (CSS) sequence, and wild type CH2 and CH3 domains. EXAMPLE 2 Culturing Host Cells with Carbohydrate modifier [0104] CHO cells transfected with TRU-016 or TRU-015 cDNA were cultured in shake flasks or wave bags with varying concentrations of various carbohydrate modifiers generally according to the procedures described below. [0105] For shake flask runs, log phase host cells were seeded in shake flasks at 100,000 cells/ml with carbohydrate modifier at the concentration to be tested, and optionally with methotrexate (MTX) @ 50 nM [0106] Cells were seeded at 3 x 10E6/mL in 1350 mL of Ex-Cell 302 culture media (SATC Biosciences; with added non-essential amino acids, pyrucate, L-glutamine, pen/strep, HT Supplement and insulin, all from Invitrogen) at t=0 and brought to 5L total volume at T>= 72 hours. The cells were incubated at 37 "C and 5% carbon dioxide and monitored for growth and viability daily starting at day 6-7. Supernatants were typically harvested at day 10-12 when cell viability dropped below 60%. [0107] Na-Azide was added to 0.02%, cells were removed by centrifugation and supernatant was filter sterilized through a 0.22 uM filter. Some assays described in other examples herein were performed on the supernatants as indicated, while other assays were performed on material that underwent further protein A purification. For wave bag runs, log phase host cells were seeded into 5L wave bags at 100,000-200,000 cells/ml in 10-20% conditioned Ex-Cell 302 media (SATC Biosciences; with added non-essential amino acids, pyrucate, L-glutamine, pen/strep, HT Supplement and insulin, all from Invitrogen) with carbohydrate modifier at the concentration to be tested. Cells were incubated at 37 "C and 5% carbon dioxide and monitored daily for growth and viability. Supernatants were typically harvested at day 11-12 or when cell viability dropped below 50%. [0108] Cells were removed by centrifugation in a Sorvall Legend at 3000 rpm (1932 rcf) for 20 minutes, the supernatant was filter sterilized. Some assays described in other 26 examples herein were performed on the supematants as indicated, while other assays were performed on material that underwent further protein A purification. [0109 TRU-016 produced by cells cultured with varying concentrations of various carbohydrate modifiers is assayed for CD16 binding, ADCC, CDC, pharmacokinetic parameters and in vivo activity as described below. [0110] Figures 1 and 2 are representative and show that treatment with the carbohydrate modifier castanospermine at concentrations up to 1000 pM did not affect cell counts or percent cell viability over all time periods sampled (up to 144 hours). EXAMPLE 3 Binding to FeRs [0111J The inimunoglycoproteins produced according to Example 2 were assayed in vitro for binding to soluble Ig-fusion versions of Fcy receptors, in which the extracellular domain of a receptor is fused to murine IgG2a Fe. 10112] The soluble Fcy receptor materials were generated by fusing the extracellular domain of Fey Receptors I (Genbank Acc. No. BC032634), Ila (Genbank Acc. No. NM_021642), 11, (Genbank Acc, No. BC03 1992), and III-V158 (high affinity allele) (Genbank Acc. No. X07934) and 11-F158 (low affinity allele), respectively, to a murine IgG2a Fe with a Pro to Ser mutation at residue 238 (MIgG2aP238S). For both forms of Fcy RII (CD16), an HE4 leader was cloned onto CD16 amino acids 1-178 and then fused to MIgG2aP238S. [01131 The assays were carried out as follows. 500,000 WIL2-S cells (a B lymphoma cell line that expresses CD37 as well as CD20 on its surface) were incubated on ice in a Costar 96 well plate with 5 jg/ml of either TRU-0 15 or TRU-0 16 for 45 minutes in phosphate buffered saline (PBS) with 1% fetal bovine serum (FBS). Unbound TRU-015 or TRU-016 was removed by spinning the cells, washing with diluent (PBS + 1 %FBS) and spinning again at 1200 rpm in a Sorvall Legend RT for 2 minutes. The cells were then incubated with the desired FcyR-MIg fusion in the same diluent at a concentration of I gg/ml on ice for 45 minutes. 27 10114] The complexes (WIL2-S cells/SMIP/ FcyR-MIg) were then incubated with PE conjugated AffiniPure F(Ab') 2 Goat Anti-Mouse IgG [Jackson Immunoresearch] (a mouse Fe-specific antibody with minimal cross reactivity with human Fc) at a 1:100 dilution. The cells were analyzed by one-color flow cytometry on a FACsCalibur using CellQuest software (Becton Dickinson). [01151 If TRU-016 supernatants from Example 2 were used in this assay instead of purified TRU-016 protein, the SMIP concentration in the supernatant was quantified by direct staining of WIL2-S cells with diluted supernatant along with a TRU-016 standard. TRU-016 was detected by staining with FITC conjugated F(Ab') 2 Goat Anti-Human (gamma) [Caltag H10101] at a 1:50 dilution. 10116] Binding to either the low affinity allele and high affinity allele were determined to correlate similarly to ADCC activity. An increase in CD 16 (low or high affinity allele) binding was correlated to an increase in ADCC activity. [0117] Representative results are displayed in Figures 3-6. [0118] TRU-015 purified protein produced by CHO cells cultured in media containing 0, 2, 5, 10, 30 or 100 gg/mL castanospermine was tested for CD16 binding (low affinity allele). Representative results of geometric mean fluorescence intensity are displayed in Figure 3 and show a dose-dependent increase in CD16 binding at increasing concentrations of castanospermine in the culture media. 101191 TRU-016 supernatant produced by CHO cells cultured in media containing 6,8a diepicastanospermine at a concentration of 50 or 250 AM, swainsonine at a concentration of 50 or 250 AM, or deoxymannojirimycin (DMJ) at a concentration of 50 or 250 pM was tested for CD16 binding. Representative results of mean fluorescence intensity are displayed in Figure 4 and show that both concentrations of DMJ increased CD16 binding. Although no effect was seen for 6
,
8 a-diepicastanospermine or swainsonine at these concentrations, further tests with purified protein are carried out to determine effect. 101201 TRU-0 16 supernatant produced by CHO cells cultured in media containing kifunensine at a concentration of 0, 0.5, 1, 3, 5, or 10 AM was tested for CD16 binding. Representative results of mean fluorescence intensity are displayed in Figure 5 and show that 28 kifunensine was much more potent than DMJ at increasing CD 16 binding and greatly increased CD16 binding even at the lowest concentration, 0.5 pM. [0121J Protein A-purified TRU-016 produced by CHO cells cultured in media containing 0, 10, 25, 50, 100 or 200 pM castanospermine was tested for CD16 binding. Representative results of mean fluorescence intensity are displayed in Figure 6 and show a dose-dependent increase in CD 16 binding at increasing concentrations of castanospermine in the culture media. EXAMPLE 4 ADCC Activity [0122] To determine the ADCC activity of purified TRU-016, labeled BJAB B cells were used as targets and human peripheral blood mononuclear cells (PBMC) as effector cells. BJAB B cells (10 7 cells) were labeled with 500 jCi/mL 5 Cr sodium chromate for 2 hours at 37 0 C in IMDM/l O%FBS. PBMCs were isolated from heparinized, human whole blood by fractionation over Lymphocyte Separation Media (LSM, ICN Biomedical) gradients. Reagent samples were added to RPMI media with 10% FBS and serial dilutions of each reagent were prepared. The 5 'Cr labeled BJAB were added at 2x10 4 cells/well. The PBMCs were then added at 5x 10 5 cells/well for a final ratio of 25:1 effectors (PBMC):targets (BJAB). Reactions were set up in quadruplicate wells of a 96 well plate. Serial dilutions of TRU-016 were added to wells at a final concentration ranging from 10 ng/mL to 20 gg/mL as indicated in the figures. Reactions were allowed to proceed for 6 hours at 37*C in 5% CO 2 prior to harvesting and counting. CPM released was measured on a Packard TopCounNXT from 50 pl dried culture supernatant. Percent specific killing was calculated by subtracting (cpm [mean of quadruplicate samples] of sample - cpm spontaneous release)/ (cpm maximal release-cpm spontaneous release) x100, and data were plotted as % specific killing versus TRU-016 concentration. [0123] Representative results are displayed in Figures 7-10. [0124] TRU-015 purified protein produced by CHO cells cultured in media containing 0, 2, 5, 10, 30 or 100 gg/mL castanospermine was tested for ADCC measured using PBMC from high affinity (V/V158) and low affinity (F/F158) CD16 donors. Representative results of % specific killing are displayed in Figures 7 and 8 (high affinity and low affinity donors, 29 respectively) and show a dose-dependent increase in ADCC activity at increasing concentrations of castanospermine in the culture media. [0125] TRU-016 purified protein produced by CHO cells cultured in media containing 0, 10, 25, 50, 100 or 200 pM castanospermine was tested for ADCC. Representative results of % specific killing are displayed in Figure 9 and show a dose-dependent increase in ADCC activity at increasing concentrations of castanospermine in the culture media. [0126] TRU-016 purified protein produced by CHO cells cultured in media containing 200 pM DMJ, 10 pM kifenunsine or 200 gM castanospermine was tested for ADCC. Representative results of % specific killing are displayed in Figure 10 and show that all of these concentrations of carbohydrate modifiers improved ADCC of the immunoglycoproteins produced by the CHO cells. EXAMPLE 5 CDC Activity [0127] To determine the CDC activity of TRU-016 purified protein produced according to Example 2, Ramos B cells were suspended in Iscoves (Gibco/lnvitrogen, Grand Island, NY) at 5x10 5 cells/well in 75 pl. TRU-016 (75 gl) were added to the cells at twice the concentrations indicated. Binding reactions were allowed to proceed for 45 minutes prior to centrifugation and washing in serum-free Iscoves. Cells were resuspended in Iscoves with human serum (containing complement) at various concentrations. The cells were incubated 60 minutes at 37"C. Cells were washed by centrifugation and resuspended in staining media with 0.5 pg/ml propidium iodide. Samples were incubated 15 minutes at room temperature in the dark prior to analysis by flow cytometry using a FACsCalibur and CellQuest software (Becton Dickinson). [01281 TRU-015 purified protein produced by untreated CHO cells, or CHO cells treated with 30 jig/ml castanospermine was tested for CDC activity. Results are displayed in Figure 15. [0129] TRU-016 purified protein produced by untreated CHO cells, or CHO cells cultured in media containing 200 gM DMJ, 10 pM kifenunsine or 200 [LM castanospermine, was tested for CDC activity. Results are displayed in Figure 16. 30 [01301 These results show that CDC for carbohydrate-modified TRU-015 or TRU-016 was similar to the CDC of corresponding protein produced by untreated CHO cells, indicating that the presence of carbohydrate modifier in the culture medium of the host cells had no significant effect on CDC of the immunoglycoprotein produced by the host cells. EXAMPLE 6 Pharmacokinetic Profile [0131] Female BALB/c mice were injected i.v. with 200 ig of TRU-016 test protein (TRU-016 produced by untreated CHO cells or by CHO cells treated with 200 pM DMJ, 10 pM kifenunsine or 200 pM castanospermine) at time 0. Serum samples were collected (3 mice per time point) at 15 min, 2, 6, 24, 48, 72, 96, and 192 hours post injection. [0132] The serum concentration of each TRU-016 test sample was determined in a FACS based binding assay using the CD37+ Ramos human cell line. CD37+ Ramos cells (5xI05 cells/well) were incubated in 96 well flat bottom plates along with the serum sample to be tested. Spiked serum samples were used for the standard curves. Cells were incubated at 4*C for an hour and washed before addition of the detection antibody. Binding of TRU-016 test protein to CD37+ Ramos cells was detected using a fluorescein-conjugated goat anti-human IgG Fc'J fragment-specific antibody. Standard curves were used to construct a binding curve as a function of antigen concentration. Briefly, standard curves consisted of various known concentrations of the TRU-016 test protein spiked into normal mouse serum diluted 1:20 in FACS buffer. The standard curves were run in duplicate on each plate. Mean fluorescence intensities (MFI) from the FACS analysis were imported into Softmax Pro software and were used to calculate serum concentrations of the TRU-016 test protein. [0133] Results of the pharmacokinetic study showed that TRU-016 produced by CHO cells cultured in media containing 200 pM DMJ, 10 pM kifenunsine or 200 pM castanospermine (displayed in Figure 11) when administered to mice exhibited a pharmacokinetic profile similar to TRU-016 produced by untreated CHO cells, indicating that carbohydrate modifier in the culture medium of the host cells had no significant effect on half-life or other pharmacokinetic parameters. [0134] Repeating the CD16 assays on sera containing TRU-016 obtained from the mice at 48, 72, 96 and 192 hours after administration of TRU-016 showed that the sera retained its increased CD16 binding activity at all time points tested. Results are shown in Figure 12. 31 EXAMPLE 7 Carbohydrate-modified immunoglycoprotein activity in vivo 10135] Nude mice are administered 5 x 106 Ramos cells subcutaneously on day 0 and injected intravenously with 200 pg control human IgG or TRU-016 test protein produced by CHO cells treated with 200 gM DMJ, 10 gM kifenunsine or 200 pM castanospermine on days 0, 2, 4, 6, and 8. Mice typically develop tumors within 6 days and die shortly thereafter. Tumors are measured three times weekly with digital calipers and LabCat software, and tumor volume is calculated as %2[length x (width)] 2 . Body weight is also determined once a week. [0136] Mice are sacrificed when the tumor reaches 1500 mm 3 in size (1200 mm 3 on Fridays). Mice are also sacrificed if ulceration of a tumor occurs, the tumor inhibits the mobility of animal, or if weight loss equals or exceeds 20%. (0137] Interim results for relative tumor volume at day 8 after the study was initiated are shown in Figure 13. Data on % survival after the initiation of study are shown in Figure 14 and below in Table 1. 32 Table 1 Group Median Survival Time p value (Days)* HuIgG 8 CS TRU-016 13 0.0054 DMJ TRU-016 13.5 0.0005 Kifu TRU-016 10 0.0084 *Values for each of the carbohydrate-modified TRU-016 are significantly different from that of the huIgG treated control group. [0138] Results of this in vivo study showed that TRU-01 6 produced by CHO cells treated with 200 pM DMJ, 10 pM kifenunsine or 200 pM castanospermine was able to reduce tumor volume and increase mean survival time in an animal model of cancer. EXAMPLE 8 Effect of Castanospermine at Varying Concentrations on Protein Production [0139] Further experiments were performed to determine the effect of castanospermine concentration on cell viability, density and specific protein production of TRU-016. [0140] Prior to initiation of the experiments, CHO cells transfected with TRU-016 were grown in shake flasks in Ex-CellTM 302 CHO serum-free media (SAFC Biosciences) supplemented with 1x non-essential amino acids (MediaTech), lx sodium pyruvate (MediaTech), 4 mM L-glutamine (MediaTech), 500 nM methotrexate (MP Biomedicals) and 1 mg/L recombinant insulin (Recombulin - GIBCO/Invitrogen Corp.) at 37 *C and 5% carbon dioxide in a humidified incubator. A 200 mM stock concentration of castanospermine (Alexis Biochemicals) was prepared by dilution of the castanospermine in sterile, distilled/deionized water (MediaTech) and filtration through a 13 mm Acrodisc® with a 0.2 pm HT Tuffryn membrane (Pall Corporation). Stock solution was aliquoted into sterile, 0 ringed, 0.5 mL microcentrifuge tubes (Fisherbrand, Fisher Scientific) and frozen at -20'C. Approximately 1 hour prior to initiation of experiments, needed aliquots were thawed at room temperature and the contents of each vial mixed well by vortexing. [0141] For each experiment, cells in log phase growth were seeded in the above medium into a total volume of 60 mL in 250 mL shaker flasks at a density of 200,000 cells/niL and 33 CS added at the concentration to be tested. Final CS concentrations of 800 pM, 400 AM, 200 AM, 100 AM, 50 AM, 25 pM and 0 pM were each tested in duplicate flasks. All cultures were incubated at 37 *C and 5% carbon dioxide in a humidified incubator and monitored at least every other day for viable cell density and overall cell viability. 101421 Cultures were harvested on day 8 when overall cell viability was 50-70% (Expt. 1) and 30-50% (Expt. 2). Cells and cellular debris were removed by centrifugation in a Sorvall Super T21 at 3000 rpm for 20 minutes after which the supernatant was sterile filtered through a Millipore Steriflip unit with a 0.22 Am Millipore Express Plus membrane and stored at 2-8 *C until purification. [0143J Although cell viability and growth did not appear to be significantly affected as indicated by each sample's integral cell area (ICA), Table 2, increasing concentrations of castanospermine appeared to reduce immunoglycoprotein production. Results are shown in Figure 17 and in Table 2 below. Concentrations of 400 pm and 800 pm CS are shown to reduce TRU-016 protein production by approximately 40%-55% respectively. [0144] Table 2 Average CS Conc. Viability TRU-016 ICA" Specific (pM) at Produced 108 cells* Productivity, Harvest (%) (ug/mL) ± SD days/mL (pg/cell/day) 800 70.6 99.65 6.1 23,9 3.99 800 65.2 23.8 4.36 400 68.2 124.93 1.4 22.4 5.53 400 65.7 23.0 5.48 200 55.5 143.53 1.4 21.9 6.60 200 51.1 22.0 6.47 100 54.4 161.83 0.1 21.6 7.48 100 54.6 21.6 7.49 50 49.4 176.63 1.0 21.6 8.15 50 50.0 21.4 8.27 25 53.9 180.31 ±6.6 21.4 8.22 25 54.5 21.1 8.78 0 65.1 208.24 0.3 22.4 9.29 0 62.6 , 21.7 9.62 Integral Cell Area (ICA) ICA = ((VCC. + VCC,,)/2) x (y-t) where VCC, = viable cell density at time n VCC,,, = viable cell density at time n+1 units: 106 cells * days/mL 34 Specific Productivity = total amount produced (ug/mL)/ICA units: pg/cell/day EXAMPLE 9 Assay for Simultaneous Binding of TRU-016 to CD37 and FeyRIIIa (CD16) [0145] Experiments were performed to determine the effect of castanospermine concentration on functional activity of TRU-016 as measured by its binding to FcyRIIIa and its binding to target antigen CD37. [01461 TRU-016 produced as described in Example 8 was tested in the following assay, which simultaneously evaluates the ability of the TRU-016 binding domain to bind to a CD37 expressing target cell and the ability of the Fc portion of the TRU-016 SMIP to bind a fusion protein of human CD16 and murine IgG Fe. 101471 The target cell utilized is the Daudi (ATCC CRL-213) cell line. Daudi cells are a human B-lymphoblastoid cell line derived from a Burkitt's lymphoma and express high levels of CD37. The custom soluble CD16:MuIgGFc fusion protein is human CD16 (low affinity polymorphism) linked to a murine IgG Fc. [0148] The appropriate number of Daudi cells (350,000/well times the number of wells) is aliquoted and centrifuged at 250 x g for 5 minutes at 15*C. The supernatant is removed. One percent cold paraformaldehyde is prepared by diluting the 4% stock from USB (USB US19943) 1:4 with FACS Buffer. FACS Buffer is prepared by adding 2% FBS (Gibco) to Dulbecco's PBS (Invitrogen) (v/v) and sterile filtering with a 0.22 gm filter. FACS Buffer is stored and used at 4*C. The cells are resuspended in 1% paraformaldehyde (a volume equal to 50 [LL/well times the number of wells) and plated out in a round bottom 96-well plate. The cells are incubated for 30 minutes at 4*C. Following this incubation the cells are washed by adding 150 gL of FACS Buffer to each well, centrifuging at 250 x g for 3 minutes at 15"C and the supernatant removed. The cells are resuspended in 50 pL of FACS Buffer. TRU-016 is diluted in FACS Buffer, at concentrations ranging from saturation to background levels (24 pg/mL - 0.011 jig/mL), added to the appropriate wells, 50 gL/well, and the cells incubated for 25 minutes at 4*C. The CD16:MuIgGFc fusion protein is diluted in FACS Buffer to a saturating level (20 pg/ml) and added to the assay (50 pL/well) and incubated for an 35 additional 30 minutes at 4*C to form a complex with the TRU-016 that has bound to the cell surface. Any unbound reagents are removed from the well by centrifuging at 250 x g for 3 minutes at I 5C, removing the supernatant and then washing 3 times with 200 AL/well of FACS Buffer. The cells are then incubated with a fluorophore (R-phycoerythrin, Jackson 115-116-071) tagged F(ab')2 antibody, specific to murine Fe (and selected to be minimally reactive to human Fc). This antibody will bind to the MuIgGFc portion of the CD16:MuIgGFc fusion protein. The antibody is diluted 1:200 in FACS Buffer and 1 0OgL is added to each well. The plate is incubated at 4*C in the dark for 45 minutes. Any unbound R-PE is removed by adding 150 jL of FACS Buffer to each well and centrifuging at 250 x g for 3 minutes at 15*C followed by removal of supernatant. This is followed by a second wash with 200 pL/well FACS Buffer, centrifuging at 250 x g for 3 minutes at 15*C and removal of supernatant. The cells are resusupended with 200 L/well 1% paraformaldehyde and stored at 4"C overnight. [01491 Each sample's bound fluorescence is measured on a BD FACSCalibur flow cytometry system and analyzed with Cell Quest Pro software (Becton Dickinson, ver 5.2). The GeoMean fluorescence intensity for each sample is plotted relative to the TRU-0 16 concentration. A dose response is generated and fit to a 4-parameter logistic (4-PL) curve using SoftMax Pro software (Molecular Devices, ver 5.0.1). Titrations of TRU-016 are utilized to create a dose response curve of test and reference material for comparison. The "D"-parameter (Maximal curve asymptote) is used as reference for comparison of treated and untreated samples. An increase in the "D" value represents in increase in the binding activity for the corresponding sample. [0150] Results of the experiment are displayed in Figure 18 and show a dose-dependent binding response relative to concentration of CS up to 400 pM, at which point the binding appears to level off. [0151] To demonstrate that the enhanced binding of CS treated TRU-016 samples to CD16 was not in part due to enhanced binding of the molecules to CD37, the above assay was repeated except that after addition and incubation of treated or untreated TRU-016 samples in the assay plate, unbound TRU-016 is removed from the well by centrifuging at 250 x g for 3 minutes at 15oC, removing the supernatant and then washing 3 times with 200 pL/well of FACS buffer. The cells are then incubated with a FITC-conjugated goat anti-human IgG Fc 36 specific antibody (Caltag H10501). This antibody will bind to the Fe region of the human IgG chain of TRU-016 bound to the cells. The antibody is diluted 1:50 in FACS buffer and 100 L is added to each well. The plate is incubated at 4oC in the dark for 45 minutes. Any unbound FITC-labeled antibody is removed by adding 100 gL of FACS buffer to each well, centrifuging at 250 x g for 3 minutes at 15oC followed by removal of supernatant. This is followed by a second wash with 200 pL/well FACS buffer. The cells are resusupended with 200 pL/well 2% paraformaldehyde and stored at 4oC overnight. Each sample's bound fluorescence is measured on a BD FACSCalibur flow cytometry system and analyzed using Cell Quest Pro software (Becton Dickinson, ver 5.2). The GeoMean fluorescence intensity for each sample is plotted relative to the TRU-0 16 concentration. A dose response curve is generated and fit to a 4-parameter logistic (4-PL) curve using the SoftMax Pro software (Molecular Devices, ver 5.0.1). Titrations ofTRU-016 are utilized to create a dose response curve of the untreated control and CS treated samples for comparison. 101521 As shown in Figure 19, the dose response binding curves to CD37 expressing cells for all CS treated samples were essentially identical to each other and to the untreated TRU 016 sample, indicating that treatment with CS did not alter the binding of TRU-016 to its specific target antigen. EXAMPLE 10 Antibody Dependent Cellular Cytotoxicity (ADCC) Assay [0153] Experiments were performed to determine the effect of castanospermine concentration on functional activity of TRU-016 as measured by ADCC activity. [01541 TRU-016 produced as described in Example 8 is incubated with the CD37 expressing Daudi cancer B-cell line in conjunction with primary human peripheral blood lymphocytes (PBL's) effector cells to assess ADCC activity. 101551 Daudi target cells (5 x 10^6) are added to a 15 ml conical tube and then centrifuged at 250 x g for 5 minutes at 20"C and the supernatant removed. The cell pellet is resuspended by the addition of 0.3 mCi Chromium-51
(
51 Cr, GE Healthcare, CJ5 1). The cells are incubated for 75 minutes at 37"C with 5% CO 2 , allowing the cells to incorporate the radioactive isotope. The cells are then washed three times to remove any unincorporated ,Cr. This is done by adding 10 mL of complete media - IMDM (Gibco) with 10% FBS (Gibco) - to the tube, centrifuging at 250 x g for 5 minutes at 20*C followed by removal of 37 supernatant. The final resuspension is in 11.5 mL of complete media. TRU-0 16 is diluted in complete media, at concentrations that are able to generate maximal to background levels of cell lysis (500 ng/mL - 0.005 ng/mL). These titrations are plated out, 50 gL/well, in a round bottom 96 well plate. The 51 Cr labeled target cells are added to the dose titrations of TRU 016 at 50 pL/well and the control wells (control media without TRU-016). PBL's are isolated from fresh heparinized whole blood by density gradient centrifugation using Lymphocyte Separation Media as per protocol (LSM, MP Biomedical, 50494/36427). PBL effector cells are added, 100 pL/well, to the wells at a ratio of between 25:1-30:1 (effector: target). The assay is incubated for 4.5 - 5 hours at 37"C, 5%CO 2 . The effector cells lyse the target cells relative to the TRU-0 16 concentration, releasing a proportional amount of 5 Cr into the assay supernatant. Following the incubation the plate is centrifuged at 250 x g for 3 minutes at 20*C. A 25 gL volume of cell-free supernatant is removed from all wells to a scintillation plate (Perkin Elmer 6005185) and dried overnight. The amount of 51 Cr isotope in each well of the scintillation plate is measured using a Topcount plate reader (Perkin Elmer, C9904VO). The data are expressed as percent of specific release. Specific release is calculated as: [0156] (Sample value - Spontaneous value) / (Maximum value - Spontaneous value) * 100% Spontaneous = amount of 1 Cr released from target cell only Maximum release = amount of 51 Cr released from targets treated with detergent lysing agent Background Control = amount of 51 Cr released from target cells + effector cells (No TRU-0 16) [0157] A dose response is generated and fit to a 4-parameter logistic curve using SoftMax Pro software (Molecular Devices, ver 5.0.1). Titrations of TRU-016 are utilized to create dose response curves of test and reference material for comparison. The EC50 values for the treated articles are compared to the untreated control (no CS) to determine the percent increase in ADCC activity. The Table below summarizes the data displayed in Figure 20. The data indicate that the ADCC activity of TRU-016, treated with CS over a range of 100 pjM - 800 pM final concentration, is significantly increased relative to untreated TRU-016. 38 Table 3 Donor Q High Donor N Low Donor AF Donor AF Sample ID 7 Heterozygous Heterozygous 1:17 1:17 1:25 1:13 Control CS OpM 1.24 2.60 0.23 0.37 CS 100pM 0.25 502%) 0.70 (370%) 0.03 (728%) n/a CS 200pM 0.21 (589% 0.54 (479% n/a 0.06 (579%) CS 400pM 0.24 (515%) 0.53 (492%) n/a 0.08 (440%) CS 800gM 0.25 (492%) 0.63 (414%) n/a 0.08 (451%) Ratio 1:X = Target to Effector (PBMC freshly isolated from whole blood) Donors are homozygous high affinity (High), homozygous low affinity (Low), or Heterozygous for CD16 allele. [01581 While the compositions and methods of this invention have been described in terms of the above-described exemplary embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims. [01591 The references cited herein throughout, to the extent that they provide exemplary details supplementary to those set forth herein, are all specifically incorporated herein by reference. 39

Claims (15)

1. A composition comprising recombinant immunoglycoproteins, wherein the recombinant immunoglycoprotein molecules each comprise a binding domain that specifically binds to CD37 and have at least a 5-fold increase in antibody-dependent cellular cytotoxicity (ADCC) as compared to control immunoglycoproteins of the same encoded amino acid sequence produced in Chinese Hamster Ovary (CHO) cells, and wherein at least 60% of the recombinant immunoglycoprotein molecules are non-fucosylated.
2. The composition of claim 1, wherein at least 80% of the recombinant immunoglycoprotein molecules are non-fucosylated.
3. The composition of claim 1, wherein at least 90% of the recombinant immu noglycoprotein molecules are non-fucosylated.
4. The composition of claim 1, wherein said recombinant immunoglycoproteins have at least a 7-fold increase in ADCC as compared to control immunoglycoproteins of the same encoded amino acid sequence produced in CHO cells.
5. The composition of claim 1, wherein said recombinant immunoglycoproteins have at least a 10-fold increase in ADCC as compared to control immunoglycoproteins of the same encoded amino acid sequence produced in CHO cells. n 4 Ina 314'21 41
6. The composition of claim 1, wherein said recombinant immunoglycoproteins are antibodies.
7. The composition of claim 1, wherein said recombinant immunoglycoproteins each comprise a binding domain that specifically binds to CD37, an immunoglobulin hinge region peptide, and immunoglobulin CH2 and CH3 domains.
8. The composition of claim 7, wherein said immunoglobulin hinge region peptide and immunoglobulin CH2 and CH3 domains are derived from a human IgG1.
9. The composition of claim 1. wherein said recombinant immunoglycoproteins each comprise a binding domain that specifically binds to CD37 and immunoglobulin CH2 and CH3 domains.
10. The composition of claim 1, wherein said recombinant immunoglycoproteins have the same serum half-life as compared to control immunoglycoproteins of the same encoded amino acid sequence produced in CHO cells.
11. The composition of claim 1 formulated in a pharmaceutically acceptable carrier or diluent.
12. A method of inhibiting cancer cell growth in a subject in need thereof comprising administering to the subject a composition of claim 1. A4 Artin 14')11- 42
13. The method of claim 12, wherein said composition is administered parenterally.
14. The method of claim 12, wherein said composition is administered by direct injection into the tissue at the site of the cancer.
15. The method of claim 12, wherein cancer cells expressing CD37 on their surface are depleted in the subject following administration of the composition. Dated this 1 st day of March 2013 Trubion Pharmaceuticals, Inc. Patent Attorneys for the Applicant PETER MAXWELL AND ASSOCIATES flA If^14 #r
AU2013201195A 2006-10-24 2013-03-01 A method for increasing antibody-dependent cytotoxicity with castanospermine Abandoned AU2013201195A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013201195A AU2013201195A1 (en) 2006-10-24 2013-03-01 A method for increasing antibody-dependent cytotoxicity with castanospermine

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/853,944 2006-10-24
AU2007308983A AU2007308983B2 (en) 2006-10-24 2007-10-24 A method for increasing antibody-dependent cytotoxicity with castanospermine
AU2013201195A AU2013201195A1 (en) 2006-10-24 2013-03-01 A method for increasing antibody-dependent cytotoxicity with castanospermine

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2007308983A Division AU2007308983B2 (en) 2006-10-24 2007-10-24 A method for increasing antibody-dependent cytotoxicity with castanospermine

Publications (1)

Publication Number Publication Date
AU2013201195A1 true AU2013201195A1 (en) 2013-03-21

Family

ID=47891163

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013201195A Abandoned AU2013201195A1 (en) 2006-10-24 2013-03-01 A method for increasing antibody-dependent cytotoxicity with castanospermine

Country Status (1)

Country Link
AU (1) AU2013201195A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3897664A4 (en) * 2018-12-19 2022-12-07 Seagen Inc. Controlled fucosylation of antibodies

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3897664A4 (en) * 2018-12-19 2022-12-07 Seagen Inc. Controlled fucosylation of antibodies

Similar Documents

Publication Publication Date Title
AU2007308983B2 (en) A method for increasing antibody-dependent cytotoxicity with castanospermine
US8383106B2 (en) Materials and methods for improved immunoglycoproteins
US20210301028A1 (en) Composition and methods for anti-tnfr2 antibodies
JP6371815B2 (en) Anti-CD19 antibodies with ADCC and CDC function and improved glycosylation profile
WO2020088605A1 (en) Homodimer-type bispecific antibody targeting cd19 and cd3, and preparation method therefor and application thereof
EP3929212A1 (en) Modified fc fragment, antibody comprising same, and application thereof
KR101364902B1 (en) Cd20 antibodies with increased fc receptor binding affinity and effector function
KR101275452B1 (en) Glycosylated antibodies
US20140286934A1 (en) Humanized antibodies that bind to cd19 and their uses
US20230340114A1 (en) Novel anti-lilrb4 antibodies and derivative products
EP2539370B1 (en) Anti-alpha2 integrin antibodies and their uses
CA2879814A1 (en) Antibodies or fusion proteins multimerized via cysteine mutation and a mu tailpiece
US20220267438A1 (en) Trispecific antibody targeting bcma, gprc5d, and cd3
AU2013201195A1 (en) A method for increasing antibody-dependent cytotoxicity with castanospermine

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application