Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 10(2): 290-303, 2018.
Article in English | MEDLINE | ID: mdl-29173063

ABSTRACT

Novel biotherapeutic glycoproteins, like recombinant monoclonal antibodies (mAbs) are widely used for the treatment of numerous diseases. The N-glycans attached to the constant region of an antibody have been demonstrated to be crucial for the biological efficacy. Even minor modifications of the N-glycan structure can dictate the potency of IgG effector functions such as the antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Here, we present the development of a glycoengineered CHO-K1 host cell line (HCL), stably expressing ß1,4-N-Acetylglucoseaminyltransferase III (GnT-III) and α-mannosidase II (Man-II), for the expression of a-fucosylated antibodies with enhanced Fc-mediated effector function. Glycoengineered HCLs were generated in a two-step strategy, starting with generating parental HCLs by stable transfection of CHO-K1 cells with GnT-III and Man-II. In a second step, parental HCLs were stably transfected a second time with these two transgenes to increase their copy number in the genetic background. Generated glycoengineered CHO-K1 cell lines expressing two different mAbs deliver antibody products with a content of more than 60% a-fucosylated glycans. In-depth analysis of the N-glycan structure revealed that the majority of the Fc-attached glycans of the obtained mAbs were of complex bisected type. Furthermore, we showed the efficient use of FcγRIIIa affinity chromatography as a novel method for the fast assessment of the mAbs a-fucosylation level. By testing different cultivation conditions for the pre-glycoengineered recombinant CHO-K1 clones, we identified key components essential for the production of a-fucosylated mAbs. The prevalent effect could be attributed to the trace element manganese, which leads to a strong increase of a-fucosylated complex- and hybrid-type glycans. In conclusion, the novel pre-glycoengineered CHO-K1 HCL can be used for the production of antibodies with high ratios of a-fucosylated Fc-attached N-glycans. Application of our newly developed FcγRIIIa affinity chromatography method during cell line development and use of optimized cultivation conditions can ultimately support the efficient development of a-fucosylated mAbs.


Subject(s)
Antibodies, Monoclonal/immunology , CHO Cells , Genetic Engineering/methods , Animals , Antibody-Dependent Cell Cytotoxicity , Cricetulus , Humans , N-Acetylglucosaminyltransferases/immunology , Protein Engineering/methods , Rats , Receptors, IgG/immunology , Transfection , alpha-Mannosidase/immunology
2.
Oncoimmunology ; 6(3): e1277306, 2017.
Article in English | MEDLINE | ID: mdl-28405498

ABSTRACT

We developed cergutuzumab amunaleukin (CEA-IL2v, RG7813), a novel monomeric CEA-targeted immunocytokine, that comprises a single IL-2 variant (IL2v) moiety with abolished CD25 binding, fused to the C-terminus of a high affinity, bivalent carcinoembryonic antigen (CEA)-specific antibody devoid of Fc-mediated effector functions. Its molecular design aims to (i) avoid preferential activation of regulatory T-cells vs. immune effector cells by removing CD25 binding; (ii) increase the therapeutic index of IL-2 therapy by (a) preferential retention at the tumor by having a lower dissociation rate from CEA-expressing cancer cells vs. IL-2R-expressing cells, (b) avoiding any FcγR-binding and Fc effector functions and (c) reduced binding to endothelial cells expressing CD25; and (iii) improve the pharmacokinetics, and thus convenience of administration, of IL-2. The crystal structure of the IL2v-IL-2Rßγ complex was determined and CEA-IL2v activity was assessed using human immune effector cells. Tumor targeting was investigated in tumor-bearing mice using 89Zr-labeled CEA-IL2v. Efficacy studies were performed in (a) syngeneic mouse models as monotherapy and combined with anti-PD-L1, and in (b) xenograft mouse models in combination with ADCC-mediating antibodies. CEA-IL2v binds to CEA with pM avidity but not to CD25, and consequently did not preferentially activate Tregs. In vivo, CEA-IL2v demonstrated superior pharmacokinetics and tumor targeting compared with a wild-type IL-2-based CEA immunocytokine (CEA-IL2wt). CEA-IL2v strongly expanded NK and CD8+ T cells, skewing the CD8+:CD4+ ratio toward CD8+ T cells both in the periphery and in the tumor, and mediated single agent efficacy in syngeneic MC38-CEA and PancO2-CEA models. Combination with trastuzumab, cetuximab and imgatuzumab, all of human IgG1 isotype, resulted in superior efficacy compared with the monotherapies alone. Combined with anti-PD-L1, CEA-IL2v mediated superior efficacy over the respective monotherapies, and over the combination with an untargeted control immunocytokine. These preclinical data support the ongoing clinical investigation of the cergutuzumab amunaleukin immunocytokine with abolished CD25 binding for the treatment of CEA-positive solid tumors in combination with PD-L1 checkpoint blockade and ADCC competent antibodies.

3.
Cancer Cell ; 31(3): 396-410, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28262554

ABSTRACT

We identified B cell maturation antigen (BCMA) as a potential therapeutic target in 778 newly diagnosed and relapsed myeloma patients. We constructed an IgG-based BCMA-T cell bispecific antibody (EM801) and showed that it increased CD3+ T cell/myeloma cell crosslinking, followed by CD4+/CD8+ T cell activation, and secretion of interferon-γ, granzyme B, and perforin. This effect is CD4 and CD8 T cell mediated. EM801 induced, at nanomolar concentrations, myeloma cell death by autologous T cells in 34 of 43 bone marrow aspirates, including those from high-risk patients and patients after multiple lines of treatment, tumor regression in six of nine mice in a myeloma xenograft model, and depletion of BCMA+ cells in cynomolgus monkeys. Pharmacokinetics and pharmacodynamics indicate weekly intravenous/subcutaneous administration.


Subject(s)
Antibodies, Bispecific/therapeutic use , B-Cell Maturation Antigen/immunology , Multiple Myeloma/drug therapy , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/pharmacology , Humans , Lymphocyte Activation , Macaca fascicularis , Mice , Xenograft Model Antitumor Assays
4.
MAbs ; 8(4): 811-27, 2016.
Article in English | MEDLINE | ID: mdl-26984378

ABSTRACT

The epidermal growth factor receptor (EGFR) and the insulin-like growth factor-1 receptor (IGF-1R) play critical roles in tumor growth, providing a strong rationale for the combined inhibition of IGF-1R and EGFR signaling in cancer therapy. We describe the design, affinity maturation, in vitro and in vivo characterization of the bispecific anti-IGF-1R/EGFR antibody XGFR*. XGFR* is based on the bispecific IgG antibody XGFR, which enabled heterodimerization of an IGF-1R binding scFab heavy chain with an EGFR-binding light and heavy chain by the "knobs-into-holes" technology. XGFR* is optimized for monovalent binding of human EGFR and IGF-1R with increased binding affinity for IGF-1R due to affinity maturation and highly improved protein stability to oxidative and thermal stress. It bears an afucosylated Fc-portion for optimal induction of antibody-dependent cell-mediated cytotoxicity (ADCC). Stable Chinese hamster ovary cell clones with production yields of 2-3 g/L were generated, allowing for large scale production of the bispecific antibody. XGFR* potently inhibits EGFR- and IGF-1R-dependent receptor phosphorylation, reduces tumor cell proliferation in cells with heterogeneous levels of IGF-1R and EGFR receptor expression and induces strong ADCC in vitro. A comparison of pancreatic and colorectal cancer lines demonstrated superior responsiveness to XGFR*-mediated signaling and tumor growth inhibition in pancreatic cancers that frequently show a high degree of IGF-1R/EGFR co-expression. XGFR* showed potent anti-tumoral efficacy in the orthotopic MiaPaCa-2 pancreatic xenograft model, resulting in nearly complete tumor growth inhibition with significant number of tumor remissions. In summary, the bispecific anti-IGF-1R/EGFR antibody XGFR* combines potent signaling and tumor growth inhibition with enhanced ADCC induction and represents a clinical development candidate for the treatment of pancreatic cancer.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/immunology , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Bispecific/biosynthesis , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cricetinae , Cricetulus , Humans , Mice , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
J Biol Chem ; 289(27): 18693-706, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24841203

ABSTRACT

In the present study, we have developed a novel one-arm single chain Fab heterodimeric bispecific IgG (OAscFab-IgG) antibody format targeting the insulin-like growth factor receptor type I (IGF-1R) and the epidermal growth factor receptor (EGFR) with one binding site for each target antigen. The bispecific antibody XGFR is based on the "knob-into-hole" technology for heavy chain heterodimerization with one heavy chain consisting of a single chain Fab to prevent wrong pairing of light chains. XGFR was produced with high expression yields and showed simultaneous binding to IGF-1R and EGFR with high affinity. Due to monovalent binding of XGFR to IGF-1R, IGF-1R internalization was strongly reduced compared with the bivalent parental antibody, leading to enhanced Fc-mediated cellular cytotoxicity. To further increase immune effector functions triggered by XGFR, the Fc portion of the bispecific antibody was glycoengineered, which resulted in strong antibody-dependent cell-mediated cytotoxicity activity. XGFR-mediated inhibition of IGF-1R and EGFR phosphorylation as well as A549 tumor cell proliferation was highly effective and was comparable with a combined treatment with EGFR (GA201) and IGF-1R (R1507) antibodies. XGFR also demonstrated potent anti-tumor efficacy in multiple mouse xenograft tumor models with a complete growth inhibition of AsPC1 human pancreatic tumors and improved survival of SCID beige mice carrying A549 human lung tumors compared with treatment with antibodies targeting either IGF-1R or EGFR. In summary, we have applied rational antibody engineering technology to develop a heterodimeric OAscFab-IgG bispecific antibody, which combines potent signaling inhibition with antibody-dependent cell-mediated cytotoxicity induction and results in superior molecular properties over two established tetravalent bispecific formats.


Subject(s)
Antibodies, Bispecific/immunology , ErbB Receptors/immunology , Immunoglobulin G/immunology , Protein Engineering , Receptor, IGF Type 1/immunology , Single-Chain Antibodies/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/metabolism , Antibodies, Bispecific/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Female , Gene Expression Regulation/drug effects , Glycosylation , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Mice , Pancreatic Neoplasms/pathology , Protein Multimerization , Protein Structure, Quaternary , Protein Transport/drug effects , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Single-Chain Antibodies/pharmacology , Xenograft Model Antitumor Assays
6.
Blood ; 115(22): 4393-402, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20194898

ABSTRACT

CD20 is an important target for the treatment of B-cell malignancies, including non-Hodgkin lymphoma as well as autoimmune disorders. B-cell depletion therapy using monoclonal antibodies against CD20, such as rituximab, has revolutionized the treatment of these disorders, greatly improving overall survival in patients. Here, we report the development of GA101 as the first Fc-engineered, type II humanized IgG1 antibody against CD20. Relative to rituximab, GA101 has increased direct and immune effector cell-mediated cytotoxicity and exhibits superior activity in cellular assays and whole blood B-cell depletion assays. In human lymphoma xenograft models, GA101 exhibits superior antitumor activity, resulting in the induction of complete tumor remission and increased overall survival. In nonhuman primates, GA101 demonstrates superior B cell-depleting activity in lymphoid tissue, including in lymph nodes and spleen. Taken together, these results provide compelling evidence for the development of GA101 as a promising new therapy for the treatment of B-cell disorders.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , B-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Humans , Immunity, Cellular , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Variable Region/genetics , In Vitro Techniques , Lymphocyte Depletion/methods , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Macaca fascicularis , Mice , Mice, SCID , Neoplasm Transplantation , Protein Engineering , Receptors, IgG/immunology , Rituximab , Transplantation, Heterologous
7.
Biotechnol Bioeng ; 93(5): 851-61, 2006 Apr 05.
Article in English | MEDLINE | ID: mdl-16435400

ABSTRACT

The effector functions elicited by IgG antibodies strongly depend on the carbohydrate moiety linked to the Fc region of the protein. Therefore several approaches have been developed to rationally manipulate these glycans and improve the biological functions of the antibody. Overexpression of recombinant beta1,4-N-acetylglucosaminyltransferase III (GnT-III) in production cell lines leads to antibodies enriched in bisected oligosaccharides. Moreover, GnT-III overexpression leads to increases in non-fucosylated and hybrid oligosaccharides. Such antibody glycovariants have increased antibody-dependent cellular cytotoxicity (ADCC). To explore a further variable besides overexpression of GnT-III, we exchanged the localization domain of GnT-III with that of other Golgi-resident enzymes. Our results indicate that chimeric GnT-III can compete even more efficiently against the endogenous core alpha1,6-fucosyltransferase (alpha1,6-FucT) and Golgi alpha-mannosidase II (ManII) leading to higher proportions of bisected non-fucosylated hybrid glycans ("Glyco-1" antibody). The co-expression of GnT-III and ManII led to a similar degree of non-fucosylation as that obtained for Glyco-1, but the majority of the oligosaccharides linked to this antibody ("Glyco-2") are of the complex type. These glycovariants feature strongly increased ADCC activity compared to the unmodified antibody, while Glyco-1 (hybrid-rich) features reduced complement-dependent cytotoxicity (CDC) compared to Glyco-2 or unmodified antibody. We show that apart from GnT-III overexpression, engineering of GnT-III localization is a versatile tool to modulate the biological activities of antibodies relevant for their therapeutic application.


Subject(s)
Immunoglobulin G/metabolism , N-Acetylgalactosaminyltransferases/genetics , Protein Engineering/methods , Protein Sorting Signals/genetics , Recombinant Fusion Proteins/metabolism , alpha-Mannosidase/genetics , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/immunology , B-Lymphocytes/immunology , CHO Cells , Cell Line , Cell Line, Tumor , Complement System Proteins/immunology , Cricetinae , Cricetulus , Cytotoxicity, Immunologic/immunology , Genetic Vectors/genetics , Glycosylation , Golgi Apparatus/enzymology , Humans , Immunoglobulin G/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Oligosaccharides/analysis , Protein Processing, Post-Translational , Receptors, IgG/metabolism , Recombinant Fusion Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...