Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Comput Aided Mol Des ; 38(1): 30, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164492

ABSTRACT

The development of novel therapeutic proteins is a lengthy and costly process, with an average attrition rate of 91% (Thomas et al. Clinical Development Success Rates and Contributing Factors 2011-2020, 2021). To increase the probability of success and ensure robust drug supply beyond approval, it is essential to assess the developability profile of new potential drug candidates as early and broadly as possible in development (Jain et al. MAbs, 2023. https://doi.org/10.1016/j.copbio.2011.06.002 ). Predicting these properties in silico is expected to be the next leap in innovation as it would enable significantly reduced development timelines combined with broader screens at lower costs. However, developing predictive algorithms typically requires substantial datasets generated under very defined conditions, a limiting factor especially for new classes of therapeutic proteins that hold immense clinical promise. Here we describe a strategy for assessing the developability of a novel class of small therapeutic Anticalin® proteins using machine learning in conjunction with a knowledge-driven approach. The knowledge-driven approach considers developability attributes such as aggregation propensity, charge variants, immunogenicity, specificity, thermal stability, hydrophobicity, and potential post-translational modifications, to calculate a holistic developability score. Based on sequence-derived descriptors as input parameters we established novel statistical models designed to predict the developability scores for Anticalin proteins. The best models yielded low root mean square errors across the entire dataset and were further validated by removing input data from individual screening campaigns and predicting developability scores for those drug candidates. The adoption of the described workflow will enable significantly streamlined preclinical development of Anticalin drug candidates and could potentially be applied to other therapeutic protein scaffolds.


Subject(s)
Computer Simulation , Machine Learning , Proteins , Humans , Proteins/chemistry , Algorithms , Drug Discovery/methods , Drug Design
2.
Metab Eng ; 20: 157-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24144501

ABSTRACT

MicroRNAs (miRNAs) are short non-coding RNAs that post-transcriptionally regulate the expression of different target genes and, thus, enable engineered gene networks to achieve complex phenotypic changes in mammalian cells. We hypothesized that exploiting this feature of miRNAs could improve therapeutic protein production processes by increasing viable cell densities and/or productivity of the mammalian cells used for manufacturing. To identify miRNAs that increase the productivity of producer cells, we performed a genome wide functional miRNA screen by transient transfection of Chinese hamster ovary (CHO) cells stably expressing an IgG1 antibody (CHO-IgG1). Using this approach, we identified nine human miRNAs that improved the productivities not only of the CHO-IgG1 cells but also of CHO cells expressing recombinant human serum albumin (HSA), demonstrating that the miRNAs act in a product-independent manner. We selected two miRNAs (miR-557 and miR-1287) positively impacting the viable cell density and the specific productivity, respectively, and then stably co-expressed them in IgG1 expressing CHO cells. In these cells, higher IgG1 titers were observed in fed-batch cultures whilst product quality was conserved, demonstrating that miRNA-based cell line engineering provides an attractive approach toward the genetic optimization of CHO producer cells for industrial applications.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Immunoglobulin G/biosynthesis , MicroRNAs , Animals , Antibodies, Monoclonal/genetics , CHO Cells , Cricetinae , Cricetulus , Genome-Wide Association Study , Humans , Immunoglobulin G/genetics , MicroRNAs/biosynthesis , MicroRNAs/genetics
3.
J Biotechnol ; 377: 13-22, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-37820750

ABSTRACT

Bispecific biotherapeutics offer potent and highly specific treatment options in oncology and immuno-oncology. However, many bispecific formats are prone to high levels of aggregation and instability, leading to prolonged development timelines, inefficient manufacturing, and high costs. The novel class of Mabcalin™ molecules consist of Anticalin® proteins fused to an IgG and are currently being evaluated in pre-clinical and clinical studies. Here, we describe a robust high-yield manufacturing platform for these therapeutic fusion proteins providing data up to commercially relevant scales. A platform upstream process was established for one of the Mabcalin bispecifics and then applied to other clinically relevant drug candidates with different IgG target specificities. Process performance was compared in 3 L bioreactors and production was scaled-up to up to 1000 L for confirmation. The Mabcalin proteins' structural and biophysical similarities enabled a downstream platform approach consisting of initial protein A capture, viral inactivation, mixed-mode anion exchange polishing, second polishing by cation exchange or hydrophobic interaction chromatography, viral filtration, buffer exchange and concentration by ultrafiltration/diafiltration. All three processes met their target specifications and achieved comparable clearance of impurities and product yields across scales. The described platform approach provides a fast and economic path to process confirmation and is well comparable to classical monoclonal antibody approaches in terms of costs and time to clinic.


Subject(s)
Antibodies, Monoclonal , Bioreactors , Antibodies, Monoclonal/chemistry , Chromatography , Ultrafiltration , Immunoglobulin G
4.
Nucleic Acids Res ; 38(12): 3999-4010, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20194116

ABSTRACT

The arrival of next-generation sequencing (NGS) technologies has led to novel opportunities for expression profiling and genome analysis by utilizing vast amounts of short read sequence data. Here, we demonstrate that expression profiling in organisms lacking any genome or transcriptome sequence information is feasible by combining Illumina's mRNA-seq technology with a novel bioinformatics pipeline that integrates assembled and annotated Chinese hamster ovary (CHO) sequences with information derived from related organisms. We applied this pipeline to the analysis of CHO cells which were chosen as a model system owing to its relevance in the production of therapeutic proteins. Specifically, we analysed CHO cells undergoing butyrate treatment which is known to affect cell cycle regulation and to increase the specific productivity of recombinant proteins. By this means, we identified sequences for >13,000 CHO genes which added sequence information of approximately 5000 novel genes to the CHO model. More than 6000 transcript sequences are predicted to be complete, as they covered >95% of the corresponding mouse orthologs. Detailed analysis of selected biological functions such as DNA replication and cell cycle control, demonstrated the potential of NGS expression profiling in organisms without extended genome sequence to improve both data quantity and quality.


Subject(s)
Gene Expression Profiling , Sequence Analysis, RNA , Animals , Butyrates/pharmacology , CHO Cells , Cricetinae , Cricetulus , DNA Repair , DNA Replication , Gene Expression/drug effects , Genes, cdc , Genomics , Mice , Rats , Recombination, Genetic , Signal Transduction , Transforming Growth Factor beta/metabolism
5.
Biotechnol Bioeng ; 105(2): 431-8, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19777598

ABSTRACT

Increase in both productivity and product yields in biopharmaceutical process development with recombinant protein producing mammalian cells can be mainly attributed to the advancements in cell line development, media, and process optimization. Only recently, genome-scale technologies enable a system-level analysis to elucidate the complex biomolecular basis of protein production in mammalian cells promising an increased process understanding and the deduction of knowledge-based approaches for further process optimization. Here, the use of gene expression profiling for the analysis of a low titer (LT) and high titer (HT) fed batch process using the same IgG producing CHO cell line was investigated. We found that gene expression (i) significantly differed in HT versus LT process conditions due to differences in applied chemically defined, serum-free media, (ii) changed over the time course of the fed batch processes, and that (iii) both metabolic pathways and 14 biological functions such as cellular growth or cell death were affected. Furthermore, detailed analysis of metabolism in a standard process format revealed the potential use of transcriptomics for rational media design as is shown for the case of lipid metabolism where the product titer could be increased by about 20% based on a lipid modified basal medium. The results demonstrate that gene expression profiling can be an important tool for mammalian biopharmaceutical process analysis and optimization.


Subject(s)
Biotechnology/methods , Cell Culture Techniques/methods , Cricetulus/genetics , Gene Expression Profiling , Animals , CHO Cells , Cricetinae , Cricetulus/metabolism
6.
J Cell Biol ; 165(6): 835-42, 2004 Jun 21.
Article in English | MEDLINE | ID: mdl-15210730

ABSTRACT

Apoptosis after growth factor withdrawal or drug treatment is associated with mitochondrial cytochrome c release and activation of Apaf-1 and caspase-9. To determine whether loss of Apaf-1, caspase-2, and caspase-9 prevented death of factor-starved cells, allowing them to proliferate when growth factor was returned, we generated IL-3-dependent myeloid lines from gene-deleted mice. Long after growth factor removal, cells lacking Apaf-1, caspase-9 or both caspase-9 and caspase-2 appeared healthy, retained intact plasma membranes, and did not expose phosphatidylserine. However, release of cytochrome c still occurred, and they failed to form clones when IL-3 was restored. Cells lacking caspase-2 alone had no survival advantage. Therefore, Apaf-1, caspase-2, and caspase-9 are not required for programmed cell death of factor-dependent cells, but merely affect its rate. In contrast, transfection with Bcl-2 provided long-term, clonogenic protection, and could act independently of the apoptosome. Unlike expression of Bcl-2, loss of Apaf-1, caspase-2, or caspase-9 would therefore be unlikely to enhance the survival of cancer cells.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Cell Survival/physiology , Proteins/physiology , Animals , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspases/deficiency , Caspases/genetics , Cell Line , Cell Survival/drug effects , Cytochromes c/analysis , Flow Cytometry , Gene Deletion , Interleukin-3/pharmacology , Mice , Mice, Knockout , Proteins/genetics , Tumor Cells, Cultured
7.
J Biotechnol ; 135(2): 217-23, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18448183

ABSTRACT

The optimization of production processes for therapeutic antibodies is a continuing challenge in pharmaceutical biotechnology. Although it could be demonstrated that vector design and host cell engineering can improve transcriptional and translational efficiency and thereby result in generation of high producer cell lines, it is not clear whether introduction of transgenes that regulate protein transport or affect post-translational modifications could further improve such industrial processes. Here, we show that heterologous expression of the transcription factor X-box binding protein-1 (XBP-1) can lead to an increase in endoplasmic reticulum (ER) content and specific therapeutic antibody productivity of Chinese hamster ovary (CHO)-DG44 cells in inoculum suspension cultures. This effect translates into 40% increased overall antibody titers in a fed-batch format where cells are grown in chemically defined serum-free media. Protein-A purified antibody products from mock-transfected cells and XBP-1 transfected cells were found to be of comparable quality with regard to glycosylation pattern and physicochemical characteristics. The data demonstrate the potential of XBP-1 engineering to improve mammalian cell culture production processes to yield high amounts of a therapeutic protein product of desired quality.


Subject(s)
Antibodies/metabolism , DNA-Binding Proteins/physiology , Immunoglobulin G/biosynthesis , Transcription Factors/physiology , Animals , Blotting, Western , CHO Cells , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cricetinae , Cricetulus , Culture Media, Serum-Free/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , X-Box Binding Protein 1
8.
J Biotechnol ; 219: 72-85, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26686315

ABSTRACT

The production of therapeutic antibodies using mammalian cells remains a high-priority in the biopharmaceutical manufacturing industry. Bioengineers have targeted different cellular processes, including transcription, translation, secretion and post-translational modifications, to overcome the metabolic bottlenecks limiting production capacity and create high-producing mammalian cell lines. The polycomb group (PcG) proteins belong to a family of chromatin regulators with important roles in multicellular development. By overexpressing and screening genes from the PcG family, we have identified an epigenetic key player for biopharmaceutical manufacturing enhancement: the transcription factor Yin Yang 1 (YY1). The overexpression of YY1 led to an increase in the production of several product genes (SEAP, VEGF165, IgG including Rituximab), provided that human YY1 (hYY1) was expressed in human cells (HeLa, HT-1080, HEK-293T, FreeStyle™ 293-F) and Chinese hamster ovary cell-derived YY1 (cYY1) was expressed in CHO cells (CHO-K1, CHO-easyC, FreeStyle™ CHO-S, CHO-B13-24, CHO-IgG1). Ectopic expression of cYY1 in the stable CHO-derived IgG producer cell lines CHO-B13-24 and CHO-IgG1 increased the antibody titer up to 6-fold, suggesting that epigenetic engineering of mammalian production cell lines could become a new strategy to improve the manufacturing of complex protein pharmaceuticals.


Subject(s)
Antibodies/metabolism , Gene Expression , Protein Engineering/methods , YY1 Transcription Factor/genetics , Animals , Antibodies/genetics , CHO Cells , Cricetulus , Epigenesis, Genetic , HeLa Cells , Humans
9.
J Biotechnol ; 120(1): 111-20, 2005 Oct 17.
Article in English | MEDLINE | ID: mdl-16043250

ABSTRACT

Production of biopharmaceuticals from mammalian cells requires generation of master, working and post-production cell banks of high quality under GMP conditions. An optimal cryopreservation strategy is needed for each new production cell line, particularly with regard to establishing production processes that are completely devoid of serum or even any animal components and to ensuring robust thaw performance for reliable production. Here, we describe a novel strategy employing flow-cytometric (FC) analysis of Annexin V-stained cells for high-throughput characterization of cell banks. Our data show that this method enables predictive evaluation of a cryopreservation strategy as early as 6h after thawing of cells. Furthermore, a broad study is presented characterizing various factors that may influence the quality of serum-free production cell banks from NSO and CHO cell lines. These results demonstrate how FC-based analysis can be used for development of future state-of-the-art cryopreservation strategies.


Subject(s)
Biopharmaceutics/methods , Cell Culture Techniques/methods , Cryopreservation/methods , Flow Cytometry/methods , Multiple Myeloma/metabolism , Pharmaceutical Preparations/metabolism , Protein Engineering/methods , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Mammals , Mice
10.
Adv Biochem Eng Biotechnol ; 127: 133-63, 2012.
Article in English | MEDLINE | ID: mdl-21290218

ABSTRACT

Development of efficient bioprocesses is essential for cost-effective manufacturing of recombinant therapeutic proteins. To achieve further process improvement and process rationalization comprehensive data analysis of both process data and phenotypic cell-level data is essential. Here, we present a framework for advanced bioprocess data analysis consisting of multivariate data analysis (MVDA), metabolic flux analysis (MFA), and pathway analysis for mapping of large-scale gene expression data sets. This data analysis platform was applied in a process development project with an IgG-producing Chinese hamster ovary (CHO) cell line in which the maximal product titer could be increased from about 5 to 8 g/L.Principal component analysis (PCA), k-means clustering, and partial least-squares (PLS) models were applied to analyze the macroscopic bioprocess data. MFA and gene expression analysis revealed intracellular information on the characteristics of high-performance cell cultivations. By MVDA, for example, correlations between several essential amino acids and the product concentration were observed. Also, a grouping into rather cell specific productivity-driven and process control-driven processes could be unraveled. By MFA, phenotypic characteristics in glycolysis, glutaminolysis, pentose phosphate pathway, citrate cycle, coupling of amino acid metabolism to citrate cycle, and in the energy yield could be identified. By gene expression analysis 247 deregulated metabolic genes were identified which are involved, inter alia, in amino acid metabolism, transport, and protein synthesis.


Subject(s)
Genomics/methods , Metabolic Flux Analysis/methods , Proteomics/methods , Statistics as Topic/methods , Animals , Gene Expression , Multivariate Analysis , Principal Component Analysis/methods
11.
J Biotechnol ; 152(4): 189-93, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21345355

ABSTRACT

DHFR-deficient CHO cells are the most commonly used host cells in the biopharmaceutical industry and over the years, individual substrains have evolved, some have been engineered with improved properties and platform technologies have been designed around them. Unexpectedly, we have observed that different DHFR-deficient CHO cells show only poor growth in fed-batch cultures even in HT supplemented medium, whereas antibody producer cells derived from these hosts achieved least 2-3 fold higher peak cell densities. Using a set of different expression vectors, we were able to show that this impaired growth performance was not due to the selection procedure possibly favouring fast growing clones, but a direct consequence of DHFR deficiency. Re-introduction of the DHFR gene reproducibly restored the growth phenotype to the level of wild-type CHO cells or even beyond which seemed to be dose-dependent. The requirement for a functional DHFR gene to achieve optimal growth under production conditions has direct implications for cell line generation since it suggests that changing to a selection system other than DHFR would require another CHO host which - especially for transgenic CHO strains and tailor-suited process platforms - this could mean significant investments and potential changes in product quality. In these cases, DHFR engineering of the current CHO-DG44 or DuxB11-based host could be an attractive alternative.


Subject(s)
Biopharmaceutics/methods , CHO Cells/physiology , Cell Proliferation/drug effects , Culture Media, Serum-Free/chemistry , Hypoxanthine/pharmacology , Tetrahydrofolate Dehydrogenase/deficiency , Thymidine/pharmacology , Animals , CHO Cells/drug effects , Cricetinae , Cricetulus , DNA Primers/genetics , Dose-Response Relationship, Drug , Gene Knockout Techniques , Polymerase Chain Reaction , Tetrahydrofolate Dehydrogenase/genetics
12.
J Biotechnol ; 146(4): 198-206, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-19958799

ABSTRACT

Genetic engineering of producer cell lines for production of therapeutic antibodies in order to increase the yield of production processes remains a continuing challenge. Recently it was shown that heterologous expression of the active, spliced form of human X-box binding protein 1 (XBP-1(s)) can increase the amount of secreted protein products in mammalian cell culture processes. However, a prerequisite for the industrial application of any cell engineering approach is the ability to generate monoclonal cell lines that stably express the engineering gene to maintain the desired phenotype. Here, we show a decrease in heterologous human XBP-1(s) expression in CHO production cells producing a therapeutic antibody product monitored over a prolonged period in serial culture. Colony formation assays (CFA) in CHO-K1 cells reveal a general survival disadvantage conferred by XBP-1(s) in this cell type. We aimed to rescue this phenotype by expressing the caspase-inhibitor XIAP (x-linked inhibitor of apoptosis). Using a set of bicistronic expression vectors we engineered an antibody producing CHO cell line with XBP-1(s) and XIAP alone and in combination. Interestingly, co-expression of both genes resulted in the highest specific productivities (Qp) and final titers in a serum-free fed-batch process in chemically defined media. Thus, the combination of secretion and anti-apoptotic engineering provides an interesting approach for future applications in industrial mammalian cell culture.


Subject(s)
Antibodies, Monoclonal/metabolism , Cell Culture Techniques/methods , DNA-Binding Proteins/biosynthesis , Protein Engineering/methods , Transcription Factors/biosynthesis , Animals , Antibodies, Monoclonal/chemistry , Apoptosis/physiology , CHO Cells , Cell Survival , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , Genomic Instability , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , X-Box Binding Protein 1 , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , X-Linked Inhibitor of Apoptosis Protein/genetics
13.
J Biotechnol ; 141(1-2): 84-90, 2009 Apr 20.
Article in English | MEDLINE | ID: mdl-19428735

ABSTRACT

Recent studies have demonstrated that the introduction of transgenes regulating protein transport or affecting post-translational modifications can further improve industrial processes for the production of therapeutic proteins in mammalian cells. Our study on improving therapeutic protein production in CHO cells by heterologous expression of the ceramide transfer protein (CERT) was initiated by the recent discovery that CERT is involved in protein kinase D (PKD)-dependent protein transport from the Golgi to the plasma membrane. We generated a set of CHO DG44 cell lines by stable integration of constructs expressing either CERT wild-type or CERT S132A, a mutant conferring increased lipid transfer activity, or a mock plasmid. CHO cells expressing heterologous CERT demonstrated significantly higher specific productivities of the therapeutic protein HSA when grown in inoculum suspension cultures. This effect translated into significantly increased overall HSA titers in a fed-batch format where cells are grown in chemically defined serum-free media. Furthermore, we could show that CERT also enhanced monoclonal antibody secretion in two IgG production cell lines with different basal productivities. The data demonstrate the potential of CERT engineering to improve mammalian cell culture production processes to yield high amounts of a therapeutic protein product of desired quality. To our knowledge, this is the first study showing a bottle neck in recombinant protein secretion at the Golgi complex in mammalian cells.


Subject(s)
Carrier Proteins/metabolism , Animals , Blotting, Western , CHO Cells , Carrier Proteins/genetics , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Albumin/genetics , Serum Albumin/metabolism
14.
J Biol Chem ; 277(1): 445-54, 2002 Jan 04.
Article in English | MEDLINE | ID: mdl-11604410

ABSTRACT

Inhibitor of apoptosis (IAP) proteins inhibit caspases, a function counteracted by IAP antagonists, insect Grim, HID, and Reaper and mammalian DIABLO/Smac. We now demonstrate that HtrA2, a mammalian homologue of the Escherichia coli heat shock-inducible protein HtrA, can bind to MIHA/XIAP, MIHB, and baculoviral OpIAP but not survivin. Although produced as a 50-kDa protein, HtrA2 is processed to yield an active serine protease with an N terminus similar to that of Grim, Reaper, HID, and DIABLO/Smac that mediates its interaction with XIAP. HtrA2 is largely membrane-associated in healthy cells, with a significant proportion observed within the mitochondria, but in response to UV irradiation, HtrA2 shifts into the cytosol, where it can interact with IAPs. HtrA2 can, like DIABLO/Smac, prevent XIAP inhibition of active caspase 3 in vitro and is able to counteract XIAP protection of mammalian NT2 cells against UV-induced cell death. The proapoptotic activity of HtrA2 in vivo involves both IAP binding and serine protease activity. Mutations of either the N-terminal alanine of mature HtrA2 essential for IAP interaction or the catalytic serine residue reduces the ability of HtrA2 to promote cell death, whereas a complete loss in proapoptotic activity is observed when both sites are mutated.


Subject(s)
Apoptosis , Microtubule-Associated Proteins , Proteins/antagonists & inhibitors , Serine Endopeptidases/physiology , Amino Acid Sequence , Binding Sites , Caspase 3 , Caspase Inhibitors , Chromosomal Proteins, Non-Histone/metabolism , Cytosol/enzymology , High-Temperature Requirement A Serine Peptidase 2 , Humans , Inhibitor of Apoptosis Proteins , Mitochondria/enzymology , Mitochondrial Proteins , Molecular Sequence Data , Neoplasm Proteins , Proteins/chemistry , Serine Endopeptidases/chemistry , Serine Endopeptidases/radiation effects , Survivin , Ultraviolet Rays , X-Linked Inhibitor of Apoptosis Protein
SELECTION OF CITATIONS
SEARCH DETAIL