Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biotechnol Prog ; 39(2): e3321, 2023 03.
Article in English | MEDLINE | ID: mdl-36546782

ABSTRACT

The COVID-19 pandemic has placed unprecedented pressure on biopharmaceutical companies to develop efficacious preventative and therapeutic treatments, which is unlikely to abate in the coming years. The importance of fast progress to clinical evaluation for treatments, which tackle unmet medical needs puts strain on traditional product development timelines, which can take years from start to finish. Although previous work has been successful in reducing phase 1 timelines for recombinant antibodies, through utilization of the latest technological advances and acceptance of greater business risk or costs, substantially faster development is likely achievable without increased risk to patients during initial clinical evaluation. To optimize lessons learned from the pandemic and maximize multi-stakeholder (i.e., patients, clinicians, companies, regulatory agencies) benefit, we conducted an industry wide benchmarking survey in September/October 2021. The aims of this survey were to: (i) benchmark current technical practices of key process and product development activities related to manufacturing of therapeutic proteins, (ii) understand the impact of changes implemented in COVID-19 accelerated Ab programs, and whether any such changes can be retained as part of sustainable long-term business practices and (iii) understand whether any accelerative action(s) taken have (negatively) impacted the wider development process. This article provides an in-depth analysis of this data, ultimately highlighting an industry perspective of how biopharmaceutical companies can sustainably adopt new approaches to therapeutic protein development and production.


Subject(s)
Biological Products , COVID-19 , Humans , Drug Industry , Biological Products/therapeutic use , Pandemics/prevention & control , Workflow
2.
Biotechnol Bioeng ; 119(3): 868-880, 2022 03.
Article in English | MEDLINE | ID: mdl-34935125

ABSTRACT

Genetically modified CHO cell lines are traditionally used for the production of biopharmaceuticals. However, an in-depth molecular understanding of the mechanism and exact position of transgene integration into the genome of pharmaceutical manufacturing cell lines is still scarce. Next-generation sequencing (NGS) holds great promise for strongly facilitating the understanding of CHO cell factories, as it has matured to a powerful and affordable technology for cellular genotype analysis. Targeted Locus Amplification (TLA) combined with NGS allows for robust detection of genomic positions of transgene integration and structural genomic changes occurring upon stable integration of expression vectors. TLA was applied to generate comparative genomic fingerprints of several CHO production cell lines expressing different monoclonal antibodies. Moreover, high producers resulting from an additional round of transfection of an existing cell line (supertransfection) were analyzed to investigate the integrity and the number of integration sites. Our analyses enabled detailed genetic characterization of the integration regions with respect to the number of integrates and structural changes of the host cell's genome. Single integration sites per clone with concatenated transgene copies could be detected and were in some cases found to be associated with genomic rearrangements, deletions or translocations. Supertransfection resulted in an increase in titer associated with an additional integration site per clone. Based on the TLA fingerprints, CHO cell lines originating from the same mother clone could clearly be distinguished. Interestingly, two CHO cell lines originating from the same mother clone were shown to differ genetically and phenotypically despite their identical TLA fingerprints. Taken together, TLA provides an accurate genetic characterization with respect to transgene integration sites compared with conventional methods and represents a valuable tool for a comprehensive evaluation of CHO production clones early in cell line development.


Subject(s)
Genome , High-Throughput Nucleotide Sequencing , Animals , CHO Cells , Cricetinae , Cricetulus , High-Throughput Nucleotide Sequencing/methods , Transgenes/genetics
3.
Biotechnol Bioeng ; 118(3): 1091-1104, 2021 03.
Article in English | MEDLINE | ID: mdl-33200817

ABSTRACT

A high degree of charge heterogeneity is an unfavorable phenomenon commonly observed for therapeutic monoclonal antibodies (mAbs). Removal of these impurities during manufacturing often comes at the cost of impaired step yields. A wide spectrum of posttranslational and chemical modifications is known to modify mAb charge. However, a deeper understanding of underlying mechanisms triggering charged species would be beneficial for the control of mAb charge variants during bioprocessing. In this study, a comprehensive analytical investigation was carried out to define the root causes and mechanisms inducing acidic variants of an immunoglobulin G1-derived mAb. Characterization of differently charged species by liquid chromatography-mass spectrometry revealed the reduction of disulfide bonds in acidic variants, which is followed by cysteinylation and glutathionylation of cysteines. Importantly, biophysical stability and integrity of the mAb are not affected. By in vitro incubation of the mAb with the reducing agent cysteine, disulfide bond degradation was directly linked to an increase of numerous acidic species. Modifying the concentrations of cysteine during the fermentation of various mAbs illustrated that redox potential is a critical aspect to consider during bioprocess development with respect to charge variant control.


Subject(s)
Antibodies, Monoclonal , Cysteine/chemistry , Disulfides/chemistry , Immunoglobulin G , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , CHO Cells , Cell Culture Techniques , Chromatography, Liquid , Cricetulus , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Immunoglobulin G/isolation & purification
4.
Biotechnol Bioeng ; 116(10): 2503-2513, 2019 10.
Article in English | MEDLINE | ID: mdl-31180133

ABSTRACT

Interleukin-2 (IL-2) is a potent molecule in cancer therapy. Clinical application, however, is limited due to its strong side effects during the treatment. We developed an IL-2 variant (IL-2v) immunocytokine to circumvent the drawbacks of the current IL-2 therapy. During the production of the IL-2v immunocytokine in Chinese hamster ovary (CHO) cells, molecules with fragmented IL-2v and therefore reduced cytokine activity can be observed. To control product fragmentation different production process conditions were investigated. By shifting temperature or pH after the cell growth phase to lower values, fragmented species can be reduced from 10% to 12% to about 4%. However, with the adopted process conditions, the effective titer is decreased concomitantly. Moreover, fermentation length and inoculation cell density are parameters to adjust fragmentation and effective titer. A suitable method for efficient process optimization is the design of experiment approach. With this procedure, novel optimal values for temperature, pH value, harvest day, and inoculation cell densities were proposed and tested subsequently. In comparison to the former process, the improved process reduces fragmentation by 66% while keeping the effective titer comparable. In summary, these findings will help to control fragmentation in CHO production processes of different IL-2v or IL-2 containing therapeutic proteins.


Subject(s)
Cell Culture Techniques , Interleukin-2/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Animals , CHO Cells , Cricetulus , Humans , Interleukin-2/genetics , Protein Stability , Recombinant Fusion Proteins/genetics
5.
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.

6.
Biotechnol Bioeng ; 112(6): 1187-99, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25545851

ABSTRACT

In-depth analytical characterization of biotherapeutics originating from different production batches is mandatory to ensure product safety and consistent molecule efficacy. Previously, we have shown unintended incorporation of tyrosine (Tyr) and leucine/isoleucine (Leu/Ile) at phenylalanine (Phe) positions in a recombinant produced monoclonal antibody (mAb) using an orthogonal MASCOT/SIEVE based approach for mass spectrometry data analysis. The misincorporation could be avoided by sufficient supply of phenylalanine throughout the process. Several non-annotated signals in the primarily chromatographic peptide separation step for apparently single Phe→Tyr sequence variants (SVs) suggest a role for isobar tyrosine isoforms. Meta- and ortho-Tyr are spontaneously generated during aerobic fed-batch production processes using Chinese hamster ovary (CHO) cell lines. Process induced meta- and ortho-Tyr but not proteinogenic para-Tyr are incorporated at Phe locations in Phe-starved CHO cultures expressing a recombinant mAb. Furthermore, meta- and ortho-Tyr are preferably misincorporated over Leu. Structural modeling of the l-phenylalanyl-tRNA-synthetase (PheRS) substrate activation site indicates a possible fit of non-cognate ortho-Tyr and meta-Tyr substrates. Dose-dependent misincorporations of Tyr isoforms support the hypothesis that meta- and ortho-Tyr are competing, alternative substrates for PheRS in CHO processes. Finally, easily accessible at-line surrogate markers for Phe→Tyr SV formation in biotherapeutic production were defined by the calculation of critical ratios for meta-Tyr/Phe and ortho-Tyr/Phe to support early prediction of SV probability, and finally, to allow for immediate process controlled Phe→Tyr SV prevention.


Subject(s)
Antibodies, Monoclonal/biosynthesis , CHO Cells/enzymology , CHO Cells/metabolism , Phenylalanine-tRNA Ligase/metabolism , Recombinant Proteins/biosynthesis , Tyrosine/metabolism , Animals , Antibodies, Monoclonal/genetics , Catalytic Domain , Cricetulus , Female , Leucine/metabolism , Models, Molecular , Phenylalanine-tRNA Ligase/chemistry , Protein Conformation , Recombinant Proteins/genetics
7.
Biotechnol Bioeng ; 111(11): 2290-302, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24890974

ABSTRACT

Mesenchymal stromal cells (MSCs) are promising candidates for cell therapy. Their therapeutic use requires extensive expansion to obtain a sufficiently high number of cells for clinical applications. State-of-the-art expansion systems, that is, primarily culture flask-based systems, are limited regarding scale-up, automation, and reproducibility. To overcome this bottleneck, microcarrier (MC)-based expansion processes have been developed. For the first time, MSCs from the perinatal sources umbilical cord (UC) and amniotic membrane (AM) were expanded on MCs. This study focuses on the comparison of flask- and Cytodex 1 MC-expanded MSCs by evaluating the influence of the expansion process on biological MSC characteristics. Furthermore, we tested the hypothesis to obtain more homogeneous MSC preparations by expanding cells on MCs in controlled large-scale bioreactors. MSCs were extensively characterized determining morphology, cell growth, surface marker expression, and functional properties such as differentiation capacity, secretion of paracrine factors, and gene expression. Based on their gene expression profile MSCs from different donors and sources clearly clustered in distinct groups solely depending on the expansion process-MC or flask culture. MC- and flask-expanded MSCs significantly differed from each other regarding surface markers and both paracrine factors and gene expression profiles. Furthermore, based on gene expression analysis, MC cultivation of MSCs in controlled bioreactor systems resulted in less heterogeneity between cells from different donors. In conclusion, MC-based MSC expansion in controlled bioreactors has the potential to reliably produce MSCs with altered characteristics and functions as compared to flask-expanded MSCs. These findings may be useful for the generation of MSCs with tailored properties for clinical applications.


Subject(s)
Bioreactors , Mesenchymal Stem Cells/physiology , Cell Culture Techniques/methods , Cell Proliferation , Gene Expression Profiling , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
8.
Cancer Cell ; 25(6): 846-59, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24898549

ABSTRACT

Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for these macrophages is macrophage colony-stimulating factor 1 (CSF-1). We generated a monoclonal antibody (RG7155) that inhibits CSF-1 receptor (CSF-1R) activation. In vitro RG7155 treatment results in cell death of CSF-1-differentiated macrophages. In animal models, CSF-1R inhibition strongly reduces F4/80(+) tumor-associated macrophages accompanied by an increase of the CD8(+)/CD4(+) T cell ratio. Administration of RG7155 to patients led to striking reductions of CSF-1R(+)CD163(+) macrophages in tumor tissues, which translated into clinical objective responses in diffuse-type giant cell tumor (Dt-GCT) patients.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Colonic Neoplasms/therapy , Macrophages/drug effects , Macrophages/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Cell Differentiation/physiology , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Cohort Studies , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Female , Humans , Macaca fascicularis , Macrophages/cytology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Models, Molecular , Receptor, Macrophage Colony-Stimulating Factor/metabolism
9.
PLoS One ; 7(7): e40328, 2012.
Article in English | MEDLINE | ID: mdl-22792284

ABSTRACT

Sequence variants in recombinant biopharmaceuticals may have a relevant and unpredictable impact on clinical safety and efficacy. Hence, their sensitive analysis is important throughout bioprocess development. The two stage analytical approach presented here provides a quick multi clone comparison of candidate production cell lines as a first stage, followed by an in-depth analysis including identification and quantitation of aberrant sequence variants of selected clones as a second stage. We show that the differential analysis is a suitable tool for sensitive and fast batch to batch comparison of recombinant proteins. The optimized approach allows for detection of not only single amino acid substitutions in unmodified peptides, but also substitutions in posttranslational modified peptides such as glycopeptides, for detection of truncated or elongated sequence variants as well as double amino acid substitutions or substitution with amino acid structural isomers within one peptide. In two case studies we were able to detect sequence variants of different origin down to a sub percentage level. One of the sequence variants (Thr → Asn) could be correlated to a cytosine to adenine substitution at DNA (desoxyribonucleic acid) level. In the second case we were able to correlate the sub percentage substitution (Phe → Tyr) to amino acid limitation in the chemically defined fermentation medium.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Software , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Base Sequence , CHO Cells , Chromatography, Gel , Cricetinae , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin G/genetics , Immunoglobulin G/isolation & purification , Molecular Sequence Data , Peptide Fragments/chemistry , Phenylalanine/genetics , Point Mutation , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Reference Standards , Sequence Analysis, DNA , Sequence Analysis, Protein/methods , Tandem Mass Spectrometry/standards , Trypsin/chemistry
10.
Nat Cell Biol ; 8(9): 1035-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16906143

ABSTRACT

Separase not only triggers anaphase of meiosis I by proteolytic cleavage of cohesin on chromosome arms, but in vitro vertebrate separase also acts as a direct inhibitor of cyclin-dependent kinase 1 (Cdk1) on liberation from the inhibitory protein, securin. Blocking separase-Cdk1 complex formation by microinjection of anti-separase antibodies prevents polar-body extrusion in vertebrate oocytes. Importantly, proper meiotic maturation is rescued by chemical inhibition of Cdk1 or expression of Cdk1-binding separase fragments lacking cohesin-cleaving activity.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Endopeptidases/metabolism , Meiosis/physiology , Oocytes/physiology , Animals , CDC2 Protein Kinase/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line , Endopeptidases/genetics , Female , Histones/metabolism , Humans , Mice , Neoplasm Proteins/metabolism , Oocytes/metabolism , Securin , Separase , Xenopus
11.
Cell Cycle ; 5(1): 11-3, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16340311

ABSTRACT

Chromosome segregation in mitosis and meiosis is triggered by activation of a large protease, separase. While it has been known for some time that the anaphase inhibitor securin regulates separase activity recent work shows that this is only half the story. In vertebrates Cdk1-dependent inhibition of separase represents a second, securin-independent branch of anaphase regulation. Furthermore, an unanticipated ability of separase to inhibit Cdk1 suggests additional, nonproteolytic functions of separase.


Subject(s)
Anaphase , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Endopeptidases/metabolism , Animals , Gene Expression Regulation, Enzymologic , Humans , Separase
12.
Mol Cell ; 19(1): 135-41, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15989971

ABSTRACT

Stable maintenance of genetic information requires chromosome segregation to occur with high accuracy. Anaphase is triggered when ring-shaped cohesin is cleaved by separase, a protease regulated by association with its inhibitor securin. Dispensability of vertebrate securin strongly suggests additional means of separase regulation. Indeed, sister chromatid separation but not securin degradation is inhibited by constitutively active cyclin-dependent kinase 1 (Cdk1) and can be rescued solely by preventing phosphorylation of separase. We demonstrate that Cdk1-dependent phosphorylation of separase is not sufficient for inhibition. In a second step, Cdk1 stably binds phosphorylated separase via its regulatory cyclin B1 subunit. Complex formation results in inhibition of both protease and kinase, and we show that vertebrate separase is a direct inhibitor of Cdk1. This unanticipated function of separase is negatively regulated by securin but independent of separase's proteolytic activity.


Subject(s)
CDC2 Protein Kinase/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/isolation & purification , Cell Extracts , Cell-Free System/metabolism , Endopeptidases/genetics , Endopeptidases/isolation & purification , Enzyme Activation , Enzyme Inhibitors/metabolism , Female , Gene Expression Regulation, Enzymologic , HeLa Cells , Humans , Mitosis , Mutation , Oocytes/chemistry , Phosphorylation , Separase , Xenopus
13.
Chromosoma ; 113(8): 409-17, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15703941

ABSTRACT

Cleavage of the ring-like cohesin complex by separase triggers segregation of sister chromatids in anaphase. This simplistic model has recently been extended by exciting discoveries on three levels: regulation of anaphase by posttranslational modifications and the cohesin protector shugoshin; non-proteolytic roles of separase; and cohesin-independent linkage of sister chromatids.


Subject(s)
Anaphase/physiology , Cell Cycle Proteins/physiology , Cytoskeletal Proteins/physiology , Endopeptidases/physiology , Animals , Chromatids/physiology , Humans , Phosphorylation , SUMO-1 Protein/physiology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/physiology , Separase
SELECTION OF CITATIONS
SEARCH DETAIL
...