Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 9457, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658627

ABSTRACT

Increased use of therapeutic monoclonal antibodies and the relatively high manufacturing costs fuel the need for more efficient production methods. Here we introduce a novel, fast, robust, and safe isolation platform for screening and isolating antibody-producing cell lines using a nanowell chip and an innovative single-cell isolation method. An anti-Her2 antibody producing CHO cell pool was used as a model. The platform; (1) Assures the single-cell origin of the production clone, (2) Detects the antibody production of individual cells and (3) Isolates and expands the individual cells based on their antibody production. Using the nanowell platform we demonstrated an 1.8-4.5 increase in anti-Her2 production by CHO cells that were screened and isolated with the nanowell platform compared to CHO cells that were not screened. This increase was also shown in Fed-Batch cultures where selected high production clones showed titers of 19-100 mg/L on harvest day, while the low producer cells did not show any detectable anti-Her2 IgG production. The screening of thousands of single cells is performed under sterile conditions and the individual cells were cultured in buffers and reagents without animal components. The time required from seeding a single cell and measuring the antibody production to fully expanded clones with increased Her-2 production was 4-6 weeks.


Subject(s)
Antibodies, Monoclonal , Cricetulus , Receptor, ErbB-2 , CHO Cells , Animals , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/biosynthesis , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , Humans , Cell Separation/methods , Single-Cell Analysis/methods
2.
Cell Rep ; 34(4): 108673, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33503414

ABSTRACT

Indoleamine 2,3-dioxygenases (IDOs) degrade l-tryptophan to kynurenines and drive the de novo synthesis of nicotinamide adenine dinucleotide. Unsurprisingly, various invertebrates, vertebrates, and even fungi produce IDO. In mammals, IDO1 also serves as a homeostatic regulator, modulating immune response to infection via local tryptophan deprivation, active catabolite production, and non-enzymatic cell signaling. Whether fungal Idos have pleiotropic functions that impact on host-fungal physiology is unclear. Here, we show that Aspergillus fumigatus possesses three ido genes that are expressed under conditions of hypoxia or tryptophan abundance. Loss of these genes results in increased fungal pathogenicity and inflammation in a mouse model of aspergillosis, driven by an alternative tryptophan degradation pathway to indole derivatives and the host aryl hydrocarbon receptor. Fungal tryptophan metabolic pathways thus cooperate with the host xenobiotic response to shape host-microbe interactions in local tissue microenvironments.


Subject(s)
Aspergillosis/physiopathology , Aspergillus fumigatus/pathogenicity , Tryptophan/metabolism , Animals , Humans , Mice
3.
Curr Pharm Biotechnol ; 21(6): 467-479, 2020.
Article in English | MEDLINE | ID: mdl-32065100

ABSTRACT

OBJECTIVE: The high cost of orphan drugs limits their access by many patients, especially in low- and middle-income countries. Many orphan drugs are off-patent without alternative generic or biosimilar versions available. Production of these drugs at the point-of-care, when feasible, could be a cost-effective alternative. METHODS: The financial feasibility of this approach was estimated by setting up a small-scale production of recombinant human acid alpha-glucosidase (rhGAA). The commercial version of rhGAA is Myozyme™, and Lumizyme™ in the United States, which is used to treat Pompe disease. The rhGAA was produced in CHO-K1 mammalian cells and purified using multiple purification steps to obtain a protein profile comparable to Myozyme™. RESULTS: The established small-scale production of rhGAA was used to obtain a realistic cost estimation for the magistral production of this biological drug. The treatment cost of rhGAA using bedside production was estimated at $3,484/gram, which is 71% lower than the commercial price of Myozyme ™. CONCLUSION: This study shows that bedside production might be a cost-effective approach to increase the access of patients to particular life-saving drugs.


Subject(s)
Glycogen Storage Disease Type II/drug therapy , Orphan Drug Production/economics , Orphan Drug Production/methods , Recombinant Proteins/isolation & purification , alpha-Glucosidases/isolation & purification , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Costs , Feasibility Studies , Glycogen Storage Disease Type II/enzymology , Humans , Recombinant Proteins/economics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , alpha-Glucosidases/economics , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
4.
Pharm Res ; 37(2): 17, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31879800

ABSTRACT

PURPOSE: Modulating sialylation of therapeutic glycoproteins may be used to influence their clearance and systemic exposure. We studied the effect of low and high sialylated IL4-10 fusion protein (IL4-10 FP) on in vitro and in vivo bioactivity and evaluated the effect of differential sialylation on pharmacokinetic parameters. METHODS: CHO cell lines producing low (IL4-10 FP lowSA) and high sialylated (IL4-10 FP highSA) fusion protein were generated. Bioactivity of the proteins was evaluated in an LPS-stimulated whole blood assay. Pharmacokinetics were studied in rats, analyzing plasma levels of IL4-10 FP upon intravenous injection. In vivo activity was assessed in an inflammatory pain mice model upon intrathecal injection. RESULTS: IL4-10 FP lowSA and IL4-10 FP highSA had similar potency in vitro. The pharmacokinetics study showed a 4-fold higher initial systemic clearance of IL4-10 FP lowSA, whereas the calculated half-life of both IL4-10 FP lowSA and IL4-10 FP highSA was 20.7 min. Finally, both IL4-10 FP glycoforms inhibited persistent inflammatory pain in mice to the same extent. CONCLUSIONS: Differential sialylation of IL4-10 fusion protein does not affect the in vitro and in vivo activity, but clearly results in a difference in systemic exposure. The rapid systemic clearance of low sialylated IL4-10 FP could be a favorable characteristic to minimize systemic exposure after administration in a local compartment.


Subject(s)
Anti-Inflammatory Agents/blood , Interleukin-10/blood , Interleukin-4/blood , N-Acetylneuraminic Acid/chemistry , Recombinant Fusion Proteins/blood , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , CHO Cells , Cricetulus , Disease Models, Animal , Glycosylation , HEK293 Cells , Humans , Interleukin-10/chemistry , Interleukin-10/pharmacology , Interleukin-4/chemistry , Interleukin-4/pharmacology , Metabolic Clearance Rate , Mice, Inbred C57BL , Pain/drug therapy , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
5.
Br J Haematol ; 180(6): 808-820, 2018 03.
Article in English | MEDLINE | ID: mdl-29468712

ABSTRACT

Based on their mechanisms-of-action, CD20 monoclonal antibodies (mAbs) are grouped into Type I [complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC)] and Type II [programmed cell death (PCD) and ADCC] mAbs. We generated 17 new hybridomas producing CD20 mAbs of different isotypes and determined unique heavy and light chain sequence pairs for 13 of them. We studied their epitope binding, binding kinetics and structural properties and investigated their predictive value for effector functions, i.e. PCD, CDC and ADCC. Peptide mapping and CD20 mutant screens revealed that 10 out of these 11 new mAbs have an overlapping epitope with the prototypic Type I mAb rituximab, albeit that distinct amino acids of the CD20 molecule contributed differently. Binding kinetics did not correlate with the striking differences in CDC activity among the mIgG2c mAbs. Interestingly, chimerization of mAb m1 resulted in a mAb displaying both Type I and II characteristics. PCD induction was lost upon introduction of a mutation in the framework of the heavy chain affecting the elbow angle, supporting that structural changes within this region can affect functional activities of CD20 mAbs. Together, these new CD20 mAbs provide further insights in the properties dictating the functional efficacy of CD20 mAbs.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/immunology , Complement System Proteins/immunology , Epitopes/immunology , Antibodies, Monoclonal, Murine-Derived/genetics , Antibody-Dependent Cell Cytotoxicity/genetics , Cell Line , Epitope Mapping , Epitopes/genetics , Humans
6.
PLoS One ; 9(8): e106025, 2014.
Article in English | MEDLINE | ID: mdl-25153680

ABSTRACT

BACKGROUND: Cow's milk-derived whey hydrolysates are nutritional substitutes for allergic infants. Safety or residual allergenicity assessment of these whey hydrolysates is crucial. Currently, rat basophilic leukemia RBL-2H3 cells expressing the human IgE receptor α-chain (huFcεRIα-RBL-2H3), sensitized with serum IgE from cow's milk allergic children, are being employed to assess in vitro residual allergenicity of these whey hydrolysates. However, limited availability and inter-lot variation of these allergic sera impede standardization of whey hydrolysate safety testing in degranulation assays. OBJECTIVE: An oligoclonal pool of chimeric human (chu)IgE antibodies against bovine ß-lactoglobulin (a major allergen in whey) was generated to increase sensitivity, specificity, and reproducibility of existing degranulation assays. METHODS: Mice were immunized with bovine ß-lactoglobulin, and subsequently the variable domains of dissimilar anti-ß-lactoglobulin mouse IgG antibodies were cloned and sequenced. Six chimeric antibodies were generated comprising mouse variable domains and human constant IgE/κ domains. RESULTS: After sensitization with this pool of anti-ß-lactoglobulin chuIgEs, huFcεRIα-expressing RBL-2H3 cells demonstrated degranulation upon cross-linking with whey, native 18 kDa ß-lactoglobulin, and 5-10 kDa whey hydrolysates, whereas a 3 kDa whey hydrolysate and cow's milk powder (mainly casein) showed no degranulation. In parallel, allergic serum IgEs were less sensitive. In addition, our pool anti-ß-lactoglobulin chuIgEs recognized multiple allergenic immunodominant regions on ß-lactoglobulin, which were also recognized by serum IgEs from cow's milk allergic children. CONCLUSION: Usage of our 'unlimited' source and well-defined pool of ß-lactoglobulin-specific recombinant chuIgEs to sensitize huFcεRIα on RBL-2H3 cells showed to be a relevant and sensitive alternative for serum IgEs from cow's milk allergic patients to assess safety of whey-based non-allergic hydrolyzed formula.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin E/immunology , Lactoglobulins/immunology , Whey/immunology , Allergens/immunology , Animals , Caseins/immunology , Cattle , Cell Line , Child , Female , Humans , Mice , Mice, Inbred BALB C , Milk Hypersensitivity/immunology , Protein Hydrolysates/immunology , Rats , Receptors, IgE/immunology , Reproducibility of Results , Whey Proteins/immunology
7.
Biotechnol Bioeng ; 106(5): 741-50, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20564612

ABSTRACT

Therapeutic monoclonal antibodies, a highly successful class of biological drugs, are conventionally manufactured in mammalian cell lines. A recent approach to increase the therapeutic effectiveness of monoclonal antibodies has been to combine two or more of them; however this increases the complexity of development and manufacture. To address this issue a method to efficiently express multiple monoclonal antibodies from a single cell has been developed and we describe here the generation of stable cell clones that express high levels of a human monoclonal antibody mixture. PER.C6 cells were transfected with a combination of plasmids containing genes encoding three different antibodies. Clones that express the three corresponding antibody specificities were identified, subcloned, and passaged in the absence of antibiotic selection pressure. At several time points, batch production runs were analyzed for stable growth and IgG production characteristics. The majority (11/12) of subclones analyzed expressed all three antibody specificities in constant ratios with total IgG productivity ranging between 15 and 20 pg/cell/day under suboptimal culture conditions after up to 67 population doublings. The growth and IgG production characteristics of the stable clones reported here resemble those of single monoclonal antibody cell lines from conventional clone generation programs. We conclude that the methodology described here is applicable to the generation of stable PER.C6(R) clones for industrial scale production of mixtures of antibodies.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Biotechnology/methods , Gene Expression , Antibodies, Monoclonal/genetics , Cell Culture Techniques , Cell Line , Genetic Vectors , Humans , Plasmids , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
9.
Transplantation ; 78(10): 1429-38, 2004 Nov 27.
Article in English | MEDLINE | ID: mdl-15599306

ABSTRACT

Clinical trials designed to achieve tolerance in humans by selectively antagonizing one of the T-cell costimulatory pathways, CD40-CD40L or CD80/CD86-CD28, are pending. However, simultaneous blockade of both pathways synergistically prevented graft rejection and successfully induced donor-specific tolerance in animal models. Synergism is also supported in human T-cells in vitro following anti-CD86 mAb and anti-CD40 mAb blockade. Therefore, in our view the most promising clinical strategy would be to antagonize both CD40 and CD86. Fast clinical entrance of this anti-CD86 and anti-CD40 bidirectional concept is highly facilitated by a single molecule approach. In the present study, a single bispecific fusion protein was constructed that specifically binds human CD40 and CD86 and which combines the antagonistic activities of both anti-CD40 and anti-CD86 humanized mAb. The anti-CD40/86 fusion protein showed tolerance inducing potential as it prevented both allogeneic T-cell expansion and generation of cytotoxic effector T cells and induced anergic antigen specific regulatory T cells. These data provide proof of concept in successfully combining the antagonistic activity of two humanized mAb with great clinical potential in transplantation and autoimmunity, in one single molecule.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, CD/immunology , CD40 Antigens/immunology , Immune Tolerance/immunology , Membrane Glycoproteins/immunology , T-Lymphocytes/immunology , B-Lymphocytes/immunology , B7-2 Antigen , Binding Sites , Cells, Cultured , Humans , Immunoglobulin Fab Fragments/immunology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Lymphocytes/immunology , Recombinant Fusion Proteins/immunology
10.
Toxicology ; 174(1): 53-65, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11972992

ABSTRACT

Monoclonal antibody (Mab) 5D12 is a potent antagonist of the CD40-CD40L pathway. This cellular interaction has been validated in a large number of experimental animal models where dys-regulation of the immune system plays a role. Chimeric 5D12 (ch5D12) was constructed to reduce the potential immunogenicity and enhance the in vivo half-life when used in humans. ch5D12 is a molecularly engineered human IgG(4) antibody containing the variable domains of the heavy and light chains of the murine version of 5D12 (mu5D12). This new chimeric Mab was tested in a marmoset experimental autoimmune encephalomyelitis model and was shown to effectively prevent disease symptoms. The results of this in vivo evaluation supported clinical use of ch5D12 for immune targeted diseases. Therefore GMP material was prepared and a GLP-compliant tissue cross-reactivity study on human tissues (3 donors/37 tissues) and cynomolgus tissues (2 donors/37 tissues) was performed. ch5D12 stained on the surface of B cells and selected dendritic cells and no unexpected cross-reactivity was observed. The identical staining patterns in human and cynomolgus tissues justified the use of cynomolgus monkeys as a relevant model for humans. A GLP-compliant safety and tolerability evaluation for ch5D12 in cynomolgus monkeys was performed using the GMP produced material. Weekly administration of ch5D12 at two dose levels for 4 weeks was shown to be safe and without any side effects in all monkeys.


Subject(s)
Antibodies, Monoclonal/toxicity , CD40 Antigens/immunology , Animals , Antibodies, Monoclonal/immunology , Autoimmunity/drug effects , Brain Chemistry/drug effects , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/immunology , Immunohistochemistry , Macaca fascicularis , Recombinant Fusion Proteins/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Species Specificity
11.
Eur J Biochem ; 269(6): 1772-9, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11895448

ABSTRACT

Bouganin is a ribosome-inactivating protein that recently was isolated from Bougainvillea spectabilis Willd. In this work, the cloning and expression of the cDNA encoding for bouganin is described. From the cDNA, the amino-acid sequence was deduced, which correlated with the primary sequence data obtained by amino-acid sequencing on the native protein. Bouganin is synthesized as a pro-peptide consisting of 305 amino acids, the first 26 of which act as a leader signal while the 29 C-terminal amino acids are cleaved during processing of the molecule. The mature protein consists of 250 amino acids. Using the cDNA sequence encoding the mature protein of 250 amino acids, a recombinant protein was expressed, purified and characterized. The recombinant molecule had similar activity in a cell-free protein synthesis assay and had comparable toxicity on living cells as compared to the isolated native bouganin.


Subject(s)
Magnoliopsida/chemistry , N-Glycosyl Hydrolases/genetics , Base Sequence , Blotting, Northern , Blotting, Western , Cell Line , Cloning, Molecular , DNA, Complementary , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Sequence Data , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/isolation & purification , Open Reading Frames , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Ribosome Inactivating Proteins , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...