Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bioengineering (Basel) ; 10(3)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36978695

ABSTRACT

The XpressCF+® cell-free protein synthesis system is a robust platform for the production of non-natural amino acids containing antibodies, which enable the site-specific conjugation of homogeneous antibody drug conjugates (ADCs) via click chemistry. Here, we present a robust and scalable means of achieving a 50-100% increase in IgG titers by combining the high productivity of cell-based protein synthesis with the unique ability of XpressCF+® reactions to produce correctly folded and assembled IgGs containing multiple non-natural amino acids at defined positions. This hybrid technology involves the pre-expression of an IgG light-chain (LC) protein in a conventional recombinant E. coli expression system, engineered to have an oxidizing cytoplasm. The prefabricated LC subunit is then added as a reagent to the cell-free protein synthesis reaction. Prefabricated LC increases IgG titers primarily by reducing the protein synthesis burden per IgG since the cell free translation machinery is only responsible for synthesizing the HC protein. Titer increases were demonstrated in four IgG products in scales ranging from 100-µL microplate reactions to 0.25-L stirred tank bioreactors. Similar titer increases with prefabricated LC were also demonstrated for a bispecific antibody in the scFvFc-FabFc format, demonstrating the generality of this approach. Prefabricated LC also increases robustness in cell-free reactions since it eliminates the need to fine-tune the HC-to-LC plasmid ratio, a critical parameter influencing IgG assembly and quality when the two IgG subunits are co-expressed in a single reaction. ADCs produced using prefabricated LC were shown to be identical to IgGs produced in cell-free alone by comparing product quality, in vitro cell killing, and FcRn receptor binding assays. This approach represents a significant step towards improving IgG titers and the robustness of cell-free protein synthesis reactions by integrating in vivo and in vitro protein production platforms.

2.
Curr Opin Biotechnol ; 76: 102719, 2022 08.
Article in English | MEDLINE | ID: mdl-35569340

ABSTRACT

Cell-free protein synthesis (CFPS) technologies have grown from lab-scale research tools to biopharmaceutical production at the Good Manufacturing Practice manufacturing scale. Multiple human clinical trials are in progress with CFPS-based products. In addition, applications of CFPS in research have continued to expand over the years and play an important role in biopharmaceutical product discovery and development. The unique, open nature of CFPS has enabled efficient non-natural amino acid (nnAA) incorporation into protein products, which expands the range of biotherapeutics that can be considered for novel treatments. The flexibility and speed of CFPS combined with novel nnAA capabilities are poised to open a new chapter in the continuing evolution of biotherapies.


Subject(s)
Biological Products , Amino Acids/chemistry , Cell-Free System/chemistry , Humans , Protein Biosynthesis , Proteins/chemistry
3.
Biotechnol Prog ; 31(3): 823-31, 2015.
Article in English | MEDLINE | ID: mdl-25826247

ABSTRACT

Cell-free protein synthesis (CFPS) systems allow for robust protein expression with easy manipulation of conditions to improve protein yield and folding. Recent technological developments have significantly increased the productivity and reduced the operating costs of CFPS systems, such that they can compete with conventional in vivo protein production platforms, while also offering new routes for the discovery and production of biotherapeutics. As cell-free systems have evolved, productivity increases have commonly been obtained by addition of components to previously designed reaction mixtures without careful re-examination of the essentiality of reagents from previous generations. Here we present a systematic sensitivity analysis of the components in a conventional Escherichia coli CFPS reaction mixture to evaluate their optimal concentrations for production of the immunoglobulin G trastuzumab. We identify eight changes to the system, which result in optimal expression of trastuzumab. We find that doubling the potassium glutamate concentration, while entirely eliminating pyruvate, coenzyme A, NAD, total tRNA, folinic acid, putrescine and ammonium glutamate, results in a highly productive cell-free system with a 95% reduction in reagent costs (excluding cell-extract, plasmid, and T7 RNA polymerase made in-house). A larger panel of other proteins was also tested and all show equivalent or improved yields with our simplified system. Furthermore, we demonstrate that all of the reagents for CFPS can be combined in a single freeze-thaw stable master mix to improve reliability and ease of use. These improvements are important for the application of the CFPS system in fields such as protein engineering, high-throughput screening, and biotherapeutics.


Subject(s)
Escherichia coli/metabolism , Immunoglobulin G/biosynthesis , Protein Biosynthesis , Protein Engineering/methods , Trastuzumab/biosynthesis , Coenzyme A/chemistry , DNA-Directed RNA Polymerases/chemistry , Escherichia coli/genetics , Gene Expression , Glutamic Acid/chemistry , Immunoglobulin G/genetics , Leucovorin/chemistry , NAD/chemistry , Polyamines/chemistry , Protein Folding , Putrescine/chemistry , Pyruvic Acid/chemistry , RNA, Transfer/chemistry , Reproducibility of Results , Trastuzumab/genetics , Viral Proteins/chemistry
4.
MAbs ; 4(2): 217-25, 2012.
Article in English | MEDLINE | ID: mdl-22377750

ABSTRACT

We describe protein synthesis, folding and assembly of antibody fragments and full-length aglycosylated antibodies using an Escherichia coli-based open cell-free synthesis (OCFS) system. We use DNA template design and high throughput screening at microliter scale to rapidly optimize production of single-chain Fv (scFv) and Fab antibody fragments that bind to human IL-23 and IL-13α1R, respectively. In addition we demonstrate production of aglycosylated immunoglobulin G (IgG 1) trastuzumab. These antibodies are produced rapidly over several hours in batch mode in standard bioreactors with linear scalable yields of hundreds of milligrams/L over a 1 million-fold change in scales up to pilot scale production. We demonstrate protein expression optimization of translation initiation region (TIR) libraries from gene synthesized linear DNA templates, optimization of the temporal assembly of a Fab from independent heavy chain and light chain plasmids and optimized expression of fully assembled trastuzumab that is equivalent to mammalian expressed material in biophysical and affinity based assays. These results illustrate how the open nature of the cell-free system can be used as a seamless antibody engineering platform from discovery to preclinical development of aglycosylated monoclonal antibodies and antibody fragments as potential therapeutics.


Subject(s)
Protein Biosynthesis , Single-Chain Antibodies/biosynthesis , Transcription, Genetic , Cell-Free System/chemistry , Glycosylation , Humans , Interleukin-13 Receptor alpha1 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-23/antagonists & inhibitors , Interleukin-23/genetics , Interleukin-23/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
5.
Methods Mol Biol ; 805: 31-41, 2012.
Article in English | MEDLINE | ID: mdl-22094798

ABSTRACT

Crude cell-free extracts are useful tools for investigating biochemical phenomena and exploiting complex enzymatic processes such as protein synthesis. Extracts derived from E. coli have been used for over 50 years to study the mechanism of protein synthesis. In addition, these S30 extracts are commonly used as a laboratory tool for protein production. The preparation of S30 extract has been streamlined over the years and now it is a relatively simple process. The procedure described here includes some suggestions for extracts to be used for ribosome display.


Subject(s)
Cell Extracts , Escherichia coli Proteins/biosynthesis , Escherichia coli/metabolism , Protein Biosynthesis , Transcription, Genetic , Escherichia coli/cytology , Escherichia coli Proteins/isolation & purification , Ribosomes/metabolism
6.
Biotechnol Bioeng ; 108(7): 1570-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21337337

ABSTRACT

Engineering robust protein production and purification of correctly folded biotherapeutic proteins in cell-based systems is often challenging due to the requirements for maintaining complex cellular networks for cell viability and the need to develop associated downstream processes that reproducibly yield biopharmaceutical products with high product quality. Here, we present an alternative Escherichia coli-based open cell-free synthesis (OCFS) system that is optimized for predictable high-yield protein synthesis and folding at any scale with straightforward downstream purification processes. We describe how the linear scalability of OCFS allows rapid process optimization of parameters affecting extract activation, gene sequence optimization, and redox folding conditions for disulfide bond formation at microliter scales. Efficient and predictable high-level protein production can then be achieved using batch processes in standard bioreactors. We show how a fully bioactive protein produced by OCFS from optimized frozen extract can be purified directly using a streamlined purification process that yields a biologically active cytokine, human granulocyte-macrophage colony-stimulating factor, produced at titers of 700 mg/L in 10 h. These results represent a milestone for in vitro protein synthesis, with potential for the cGMP production of disulfide-bonded biotherapeutic proteins.


Subject(s)
Biotechnology/methods , Escherichia coli/enzymology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Technology, Pharmaceutical/methods , Bioreactors , Cell-Free System , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
7.
Biotechnol Bioeng ; 94(4): 618-24, 2006 Jul 05.
Article in English | MEDLINE | ID: mdl-16673418

ABSTRACT

Cell-free protein synthesis is a useful research tool and now stands poised to compete with in vivo expression for commercial production of proteins. However, both the extract preparation and protein synthesis procedures must be scaled up. A key challenge is producing the required amount of biomass that also results in highly active cell-free extracts. In this work, we show that the growth rate of the culture dramatically affects extract performance. Extracts prepared from cultures with a specific growth rate of 0.7/h or higher produced approximately 0.9 mg/mL of chloramphenicol acetyl transferase (CAT) in a batch reaction. In contrast, when the source culture growth rate was 0.3/h, the resulting extract produced only 0.5 mg/mL CAT. Examination of the ribosome content in the extracts revealed that the growth rate of the source cells strongly influenced the final ribosome concentration. Polysome analysis of cell-free protein synthesis reactions indicated that about 22% of the total 70S ribosomes are in polysomes for all extracts regardless of growth rate. Furthermore, the overall specific production from the 70S ribosomes is about 22 CAT proteins per ribosome over the course of the reaction in all cases. It appears that rapid culture growth rates are essential for producing a productive extract. However, growth rate does not seem to influence specific ribosome activity. Rather, the increase in extract productivity is a result of a higher ribosome concentration. These results are important for cell-free technology and also suggest an assay for intrinsic in vivo protein synthesis activity.


Subject(s)
Chloramphenicol O-Acetyltransferase/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Cell-Free System , Centrifugation, Density Gradient , Culture Media , Fermentation , Kinetics , Proteins/isolation & purification
8.
Biotechnol Prog ; 21(2): 460-5, 2005.
Article in English | MEDLINE | ID: mdl-15801786

ABSTRACT

Escherichia coli extracts activate cell-free protein synthesis systems by providing the catalysts for translation and other supporting reactions. Recent results suggest that high-density fermentations can be used to provide the source cells, but the subsequent cell extract preparation procedure requires multiple centrifugation and dialysis steps as well as an expensive runoff reaction. In the work reported here, the extract preparation protocol duration was reduced by nearly 50% by significantly shortening several steps. In addition, by optimizing the runoff incubation, overall reagent costs were reduced by 70%. Nonetheless, extracts produced from the shorter, less expensive procedure were equally active. Crucial steps were further examined to indicate minimal ribosome loss during the standard 30,000g centrifugations. Furthermore, sucrose density centrifugation analysis indicated that although an incubation step significantly activates the extract, ribosome/polysome dissociation is not required. These insights suggest that consistent cell extract can be produced more quickly and with considerably less expense for large-scale cell-free protein production, especially when combined with high-density fermentation protocols.


Subject(s)
Escherichia coli/genetics , Recombinant Proteins/biosynthesis , Cell-Free System , Centrifugation , Electrophoresis, Polyacrylamide Gel , Fermentation
9.
Biotechnol Bioeng ; 89(4): 407-15, 2005 Feb 20.
Article in English | MEDLINE | ID: mdl-15635610

ABSTRACT

A novel feeding strategy that prolongs rapid growth rates for Escherichia coli fermentations to moderately high cell density is presented. High-density fermentations are a common and successful means of producing biological products. However, acetate accumulation can be a substantial problem in these procedures. To avoid this problem, many feeding strategies and host modifications have been developed, but all result in relatively low growth rates. If a faster growth rate could be maintained, the growth phase of the process would be shortened, leading to increased productivity. It is also possible that the subsequent specific production rate could be enhanced by growing the early culture at a faster rate. We have developed a procedure to enable rapid growth to a cell density of 20 g/L and have used cell-free protein synthesis to evaluate the relative potential of the resulting cells for producing recombinant proteins. The method uses glucose pulses and the duration of the dissolved oxygen response to calculate the appropriate glucose feed rate based on the glucose demand of the culture. Amino acids and vitamins were supplied in the medium to increase the growth rate. We were able to sustain a growth rate of 0.8/h up to 20 g/L dry cell weight without significant acetate accumulation. Analysis of amino acid consumption indicates that cell composition is an accurate predictor of amino acid demand for most amino acids. Cell-free protein synthesis was used to compare the protein production potential of the high-density cultures with that of cells grown in complex medium and harvested at low cell density and maximum growth rate. Protein production for the extract from the controlled, high-density fermentations was 950 mg/L compared with 860 mg/L for the low-density control. Therefore, the new control procedure has promising potential for developing rapid and productive industrial fermentations.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/growth & development , Industrial Microbiology/methods , Acetic Acid/analysis , Amino Acids/metabolism , Fermentation , Glucose/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...