Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Methods ; 154: 38-50, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30366098

ABSTRACT

Bispecific monoclonal antibodies can bind two protein targets simultaneously and enable therapeutic modalities inaccessible by traditional mAbs. Bispecific formats containing a heterodimeric Fc region are of particular interest, as a heterodimeric Fc empowers both bispecificity and altered valencies while retaining the developability and druggability of a monoclonal antibody. We present a robust heterodimeric Fc platform, called the XmAb® bispecific platform, engineered for efficient development of bispecific antibodies and Fc fusions of multiple formats. First, we engineer a purification solution for proteins containing a heterodimeric Fc using engineered isoelectric point differences in the Fc region that enable straightforward purification of the heterodimeric species. Then, we combine this purification solution with a novel set of Fc substitutions capable of achieving heterodimer yields over 95% with little change in thermostability. Next, we illustrate the flexibility of our heterodimeric Fc with a case study in which a wide range of tumor-associated antigen × CD3 bispecifics are generated, differing in choice of tumor antigen, affinities for both tumor antigen and CD3, and tumor antigen valency. Finally, we present manufacturing data reinforcing the robustness of the heterodimeric Fc platform at scale.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal , Protein Engineering/methods , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Humans
2.
Blood ; 119(9): 2074-82, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22246035

ABSTRACT

HM1.24, an immunologic target for multiple myeloma (MM) cells, has not been effectively targeted with therapeutic monoclonal antibodies (mAbs). In this study, we investigated in vitro and in vivo anti-MM activities of XmAb5592, a humanized anti-HM1.24 mAb with Fc-domain engineered to significantly enhance FcγR binding and associated immune effector functions. XmAb5592 increased antibody-dependent cellular cytotoxicity (ADCC) several fold relative to the anti-HM1.24 IgG1 analog against both MM cell lines and primary patient myeloma cells. XmAb5592 also augmented antibody dependent cellular phagocytosis (ADCP) by macrophages. Natural killer (NK) cells became more activated by XmAb5592 than the IgG1 analog, evidenced by increased cell surface expression of granzyme B-dependent CD107a and MM cell lysis, even in the presence of bone marrow stromal cells. XmAb5592 potently inhibited tumor growth in mice bearing human MM xenografts via FcγR-dependent mechanisms, and was significantly more effective than the IgG1 analog. Lenalidomide synergistically enhanced in vitro ADCC against MM cells and in vivo tumor inhibition induced by XmAb5592. A single dose of 20 mg/kg XmAb5592 effectively depleted both blood and bone marrow plasma cells in cynomolgus monkeys. These results support clinical development of XmAb5592, both as a monotherapy and in combination with lenalidomide, to improve patient outcome of MM.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antigens, CD/immunology , Immunoglobulin Fc Fragments/immunology , Multiple Myeloma/therapy , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Degranulation , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Drug Synergism , Female , GPI-Linked Proteins/immunology , Humans , Killer Cells, Natural/immunology , Lenalidomide , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Depletion , Macaca fascicularis , Mice , Mice, SCID , Phagocytosis/drug effects , Phagocytosis/immunology , Plasma Cells/drug effects , Plasma Cells/immunology , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Xenograft Model Antitumor Assays
3.
MAbs ; 3(6): 546-57, 2011.
Article in English | MEDLINE | ID: mdl-22123055

ABSTRACT

Bispecific antibodies based on full-length antibody structures are more optimal than fragment-based formats because they benefit from the favorable properties of the Fc region. However, the homodimeric nature of Fc effectively imposes bivalent binding on all current full-length bispecific antibodies, an attribute that can result in nonspecific activation of cross-linked receptors. We engineered a novel bispecific format, referred to as mAb-Fv, that utilizes a heterodimeric Fc region to enable monovalent co-engagement of a second target antigen in a full-length context. mAb-Fv constructs co-targeting CD16 and CD3 were expressed and purified as heterodimeric species, bound selectively to their co-target antigens, and mediated potent cytotoxic activity by NK cells and T cells, respectively. The capacity to co-engage distinct target antigens simultaneously with different valencies is an improved feature for bispecific antibodies with promising therapeutic implications.


Subject(s)
Antibodies, Bispecific/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , CD3 Complex/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fragments/immunology , Receptors, IgG/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , CD3 Complex/genetics , CD3 Complex/metabolism , Dimerization , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Killer Cells, Natural/immunology , Mice , Models, Molecular , Protein Engineering/methods , Receptors, IgG/genetics , Receptors, IgG/metabolism , T-Lymphocytes/immunology
4.
J Mol Biol ; 396(5): 1474-90, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20045416

ABSTRACT

Fully human monoclonal antibodies (mAbs) derived from transgenic mice or human antibody libraries are the current state of the art for reducing the immunogenicity risk of antibody drugs. Here, we describe a novel method for generating fully human mAbs from nonhuman variable regions using information from the human germline repertoire. Central to our strategy is the rational engineering of residues within and proximal to CDRs and the V(H)/V(L) interface by iteratively exploring substitutions to the closest human germline sequences using semi-automated computational methods. Starting from the parent murine variable regions of three currently marketed mAbs targeting CD25, vascular endothelial growth factor, and tumor necrosis factor alpha, we have generated fully human antibodies with 59, 46, and 45 substitutions, respectively, compared to the parent murine sequences. A large number of these substitutions were in the CDRs, which are typically avoided in humanization methods. Antigen affinities of the fully human variants were comparable to the chimeric mAbs in each case. Furthermore, in vitro functional characterization indicated that all retain potency of the chimeric mAbs and have comparable activity to their respective marketed drugs daclizumab, bevacizumab, and infliximab. Based on local and global sequence identity, the sequences of our engineered mAbs are indistinguishable from those of fully human mAbs isolated from transgenic mice or human antibody libraries. This work establishes a simple rational engineering methodology for generating fully human antibody therapeutics from murine mAbs produced from standard hybridoma technology.


Subject(s)
Antibodies, Monoclonal/genetics , Immunoglobulin Variable Region/genetics , Protein Engineering/methods , Amino Acid Sequence , Amino Acid Substitution , Animals , Cells, Cultured , Complementarity Determining Regions/genetics , Humans , In Vitro Techniques , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Mice , Mice, Transgenic , Molecular Sequence Data , Peptide Library , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Homology, Amino Acid , Species Specificity , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
5.
J Immunol ; 179(3): 1872-83, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641054

ABSTRACT

TNF is a pleiotropic cytokine required for normal development and function of the immune system; however, TNF overexpression also induces inflammation and is associated with autoimmune diseases. TNF exists as both a soluble and a transmembrane protein. Genetic studies in mice have suggested that inflammation in disease models involves soluble TNF (solTNF) and that maintenance of innate immune function involves transmembrane TNF (tmTNF). These findings imply that selective pharmacologic inhibition of solTNF may be anti-inflammatory and yet preserve innate immunity to infection. To address this hypothesis, we now describe dominant-negative inhibitors of TNF (DN-TNFs) as a new class of biologics that selectively inhibits solTNF. DN-TNFs blocked solTNF activity in human and mouse cells, a human blood cytokine release assay, and two mouse arthritis models. In contrast, DN-TNFs neither inhibited the activity of human or mouse tmTNF nor suppressed innate immunity to Listeria infection in mice. These results establish DN-TNFs as the first selective inhibitors of solTNF, demonstrate that inflammation in mouse arthritis models is primarily driven by solTNF, and suggest that the maintenance of tmTNF activity may improve the therapeutic index of future anti-inflammatory agents.


Subject(s)
Arthritis, Experimental/immunology , Immunity, Innate , Inflammation Mediators/physiology , Listeriosis/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor , Female , Humans , Immunity, Innate/genetics , Inflammation Mediators/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/blood , Interleukin-8/metabolism , Listeriosis/genetics , Listeriosis/pathology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Paracrine Communication/immunology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Solubility , Tumor Necrosis Factor-alpha/genetics , U937 Cells
6.
Science ; 301(5641): 1895-8, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-14512626

ABSTRACT

Tumor necrosis factor (TNF) is a key regulator of inflammatory responses and has been implicated in many pathological conditions. We used structure-based design to engineer variant TNF proteins that rapidly form heterotrimers with native TNF to give complexes that neither bind to nor stimulate signaling through TNF receptors. Thus, TNF is inactivated by sequestration. Dominant-negative TNFs represent a possible approach to anti-inflammatory biotherapeutics, and experiments in animal models show that the strategy can attenuate TNF-mediated pathology. Similar rational design could be used to engineer inhibitors of additional TNF superfamily cytokines as well as other multimeric ligands.


Subject(s)
Protein Engineering , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Substitution , Animals , Antigens, CD/metabolism , Apoptosis , Arthritis, Experimental/drug therapy , Biopolymers , Caspases/metabolism , Cell Line , Cell Nucleus/metabolism , Computer Simulation , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Galactosamine/pharmacology , HeLa Cells , Humans , Liver/drug effects , NF-kappa B/metabolism , Point Mutation , Rats , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Transcription Factor RelA , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...